1
|
Wang X, Liu Z, Lin C. Metal ions-induced programmed cell death: how does oxidative stress regulate cell death? Life Sci 2025; 374:123688. [PMID: 40328371 DOI: 10.1016/j.lfs.2025.123688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 04/20/2025] [Accepted: 05/01/2025] [Indexed: 05/08/2025]
Abstract
In recent years, the mechanisms of ferroptosis and cuproptosis, two novel modes of cell death, have been elucidated and have attracted much attention. Ferroptosis is dependent on the metabolic disruption of iron ions and lipid peroxidation, whereas cuproptosis is closely related to intracellular accumulation of copper ions, aggregation of lipoylated proteins and damage to FeS cluster proteins. In particular, oxidative stress plays an important role in both types of cell death. During ferroptosis, the central role of oxidative stress is reflected in the overproduction of reactive oxygen species (ROS) and lipid peroxidation of the cell membrane. Recent studies have revealed that ROS can propagate over long distances across cells in the form of trigger waves, triggering large-scale ferroptosis. In embryonic development, different regional redox states can limit the long-distance propagation of ferroptosis waves, which is critical for muscle remodeling and tissue formation during development. In cuproptosis, processes such as copper ions accumulation, tricarboxylic acid (TCA) cycle blockade, and reduced level of FeS cluster proteins are closely associated with oxidative stress. In addition, there is a close link between oxidative stress and death induced by other metal ions (Ca2+, Zn2+, etc.). In this paper, we review the role of oxidative stress in ferroptosis and cuproptosis and the related research progress to provide new ideas for understanding the mechanism of cell death and the occurrence and treatment of related diseases.
Collapse
Affiliation(s)
- Xingsheng Wang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Zuohao Liu
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Changjun Lin
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
2
|
Yu L, Tian D, Su Z, Zhang L, Jie L, Guo S, Zhu W, Zhang N, Wang P. Mechanical stress overload promotes NF-κB/NLRP3-mediated osteoarthritis synovitis and fibrosis through Piezo1. Cell Signal 2025; 132:111786. [PMID: 40221068 DOI: 10.1016/j.cellsig.2025.111786] [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: 02/07/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025]
Abstract
Mechanical stress is a pivotal factor in the development of knee osteoarthritis (KOA). Piezo1, an innovative mechanosensitive ion channel, plays a key role in detecting variations in mechanical stress and transforming them into electrical signals. This research focuses on examining how Piezo1 influences synovial inflammation and fibrosis induced by mechanical stress in KOA, as well as delving into the potential underlying mechanisms. In vivo, pathological changes and immunohistochemical staining were conducted on both normal and overexercise rat synovial tissues to analyze the expression of Piezo1 and the NF-κB/NLRP3 pathways. In vitro utilized a cell stretcher to replicate the mechanical conditions seen in KOA. Levels of pro-inflammatory cytokines and fibrosis-related markers were assessed to investigate the impact of Piezo1 on mechanical stress in fibroblast-like synoviocytes (FLS). Subsequently, following cell stretching interventions, the effects on synovial inflammation and fibrosis were observed with the use of the Piezo1 inhibitor GsMTx4 or the NLRP3 inhibitor MCC950. Mechanical stress significantly promoted the activation of Piezo1, increased the phosphorylation ratio of p65, and elevated the levels of NLRP3, caspase-1, ASC, GSDMD, IL-1β, IL-18, IL-6, and TNF-α. Both in vitro and in vivo, mechanical stress also promoted the occurrence and development of synovial fibrosis, with significant increases in the expression levels of fibrosis-related markers. Under mechanical stress overload, upregulation of Piezo1 can promote the secretion of pro-inflammatory cytokines and the fibrotic process in synovium through the NF-κB/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Likai Yu
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Di Tian
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Zishan Su
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Li Zhang
- Orthopedics of traditional Chinese Medicine, Zhongda Hospital Southeast University, Nanjing 210009, Jiangsu, China
| | - Lishi Jie
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Shaobo Guo
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Wenhui Zhu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Nongshan Zhang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China.
| | - Peimin Wang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
3
|
Guo W, Duan Z, Wu J, Zhou BP. Epithelial-mesenchymal transition promotes metabolic reprogramming to suppress ferroptosis. Semin Cancer Biol 2025; 112:20-35. [PMID: 40058616 DOI: 10.1016/j.semcancer.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular de-differentiation process that provides cells with the increased plasticity and stem cell-like traits required during embryonic development, tissue remodeling, wound healing and metastasis. Morphologically, EMT confers tumor cells with fibroblast-like properties that lead to the rearrangement of cytoskeleton (loss of stiffness) and decrease of membrane rigidity by incorporating high level of poly-unsaturated fatty acids (PUFA) in their phospholipid membrane. Although large amounts of PUFA in membrane reduces rigidity and offers capabilities for tumor cells with the unbridled ability to stretch, bend and twist in metastasis, these PUFA are highly susceptible to lipid peroxidation, which leads to the breakdown of membrane integrity and, ultimately results in ferroptosis. To escape the ferroptotic risk, EMT also triggers the rewiring of metabolic program, particularly in lipid metabolism, to enforce the epigenetic regulation of EMT and mitigate the potential damages from ferroptosis. Thus, the interplay among EMT, lipid metabolism, and ferroptosis highlights a new layer of intricated regulation in cancer biology and metastasis. Here we summarize the latest findings and discuss these mutual interactions. Finally, we provide perspectives of how these interplays contribute to cellular plasticity and ferroptosis resistance in metastatic tumor cells that can be explored for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Wenzheng Guo
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Zhibing Duan
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Jingjing Wu
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Binhua P Zhou
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
4
|
Ke A, Yang W, Zhang W, Chen Y, Meng X, Liu J, Dai D. The cardiac glycoside periplocymarin sensitizes gastric cancer to ferroptosis via the ATP1A1-Src-YAP/TAZ-TFRC axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156804. [PMID: 40311597 DOI: 10.1016/j.phymed.2025.156804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Targeting ferroptosis vulnerabilities in tumors has become an increasingly promising therapeutic strategy. While the regulatory effects of natural products on ferroptosis are progressively being elucidated, the role of cardiac glycosides in modulating ferroptosis remains poorly understood. PURPOSE This study aims to investigate the ferroptosis-sensitizing effects of periplocymarin (PPM), a cardiac glycoside derived from the traditional plant Periploca sepium, and to elucidate the underlying molecular mechanisms. METHODS The effects of PPM on ferroptosis regulation were comprehensively assessed through functional assays, followed by sequencing analysis to identify associated signaling pathways. Subsequent mechanistic validation experiments were conducted to confirm the upstream and downstream regulatory components involved in this ferroptosis-modulating axis. RESULTS PPM induced slow and mild apoptosis in gastric cancer cells through the inhibition of glycolysis. However, when combined with ferroptosis inducers, it promoted rapid and robust ferroptosis. In vivo, PPM sensitized gastric cancer xenografts to cisplatin-induced ferroptosis with no observable cardiotoxicity or renal impairment. Mechanistically, PPM targeted the α1 subunit of the Na+/K+-ATPase (ATP1A1), leading to the activation of Src, which subsequently induced tyrosine phosphorylation of YAP/TAZ in a Hippo-independent manner, promoting their nuclear translocation. The YAP/TAZ-TEAD transcriptional complex directly bound to the TFRC promoter region between nucleotides 401-409 upstream of the transcription start site, thereby activating TFRC transcription. This resulted in increased iron influx, elevated lipid peroxidation, and heightened sensitivity to ferroptosis. Notably, ATP1A1 was essential for ferroptosis resistance, as its knockdown mimicked the sensitizing effect of PPM on ferroptosis. Moreover, the oncogenic Src-YAP/TAZ-TFRC axis may have represented a ferroptosis vulnerability and a potential biomarker in ferroptosis therapy for cancer. Importantly, other cardiac glycosides targeting Na+/K+-ATPase, such as digitoxin and bufalin, also enhanced ferroptosis sensitivity in gastric cancer cells through activation of YAP/TAZ signaling. CONCLUSION Our findings establish the cardiac glycoside PPM as a novel ferroptosis sensitizer that targets ATP1A1 to activate the Src-YAP/TAZ-TFRC axis, providing mechanistic insights for repurposing cardiac glycosides as ferroptosis modulators in precision combinatorial cancer therapy.
Collapse
Affiliation(s)
- Angting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Weiguang Yang
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wanchuan Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yibin Chen
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xiangyu Meng
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Jie Liu
- Translational Research Experiment Department, Science Experiment Center, China Medical University, Shenyang 110122, China
| | - Dongqiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
5
|
Li Q, Li C, Liu X, Guo Z, Li X, Zhang X. The key role of Piezo1 channels in ferroptosis after spinal cord injury and the therapeutic potential of Piezo1 inhibitors. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:132-140. [PMID: 40339662 DOI: 10.1016/j.pbiomolbio.2025.05.001] [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: 02/03/2025] [Revised: 04/07/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Ferroptosis has been confirmed to be one of the key mechanisms of neuronal injury and dysfunction after spinal cord injury (SCI). Mechanical stresses such as deformation, compression, and stretching not only directly cause physical damage to spinal cord tissue at the moment of SCI, but also promote the development of ferroptosis through various pathways. However, the mechanism of ferroptosis after SCI remains unclear, which hinders the development of therapeutic methods. OBJECTIVE This article aims to review the key mechanisms by which mechanical stress affects ferroptosis after SCI, including its impact on the structure and function of the endoplasmic reticulum (ER) and mitochondria, its role in triggering inflammatory responses, and its activation of mechanosensitive channels. Special emphasis is placed on the role of Piezo1 channels, which are key factors in cell mechanosensation and ion homeostasis regulation. The review explores how Piezo1 channels are upregulated by mechanical stress after SCI and participate in the ferroptosis process by mediating ion flow and other mechanisms. CONCLUSIONS Inhibiting Piezo1 channels may be a potential therapeutic strategy for SCI. This review summarizes the therapeutic potential of Piezo1 inhibitors by sorting out existing studies, hoping to provide a theoretical basis for effective therapeutic strategies targeting ferroptosis after SCI.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinyu Liu
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Zixuan Guo
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinxin Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
6
|
Han L, Tian X, Yang X, Li T, Wang S, Bao Y, Meng X. The pathogenesis of hepatocellular carcinoma: ERK/ULK1/NCOA4-mediated inhibition of iron autophagy, and Epimedium extract targeted modulation of this pathway to treat hepatocellular carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156666. [PMID: 40121885 DOI: 10.1016/j.phymed.2025.156666] [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: 10/11/2024] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND The pathogenesis of hepatocellular carcinoma (HCC) is characterized by its complexity and diversity, involving processes such as glycolysis, autophagy, and cellular immunity. Notably, the role of ERK/ULK1/NCOA4-mediated inhibition of iron autophagy in HCC pathogenesis has not been previously reported. This study provides a novel elucidation of HCC pathogenesis and identifies the clinical adjuvant therapy drug, Epimedium, as a potential treatment based on this mechanism. The research clarifies the regulatory effects of Epimedium on the ERK/ULK1/NCOA4-mediated inhibition of iron autophagy pathway in the treatment of HCC, thereby offering a scientific foundation for clinical treatment strategies and the development of innovative drugs. PURPOSE The objective of this study is to uncover a new aspect of HCC pathogenesis, ERK/ULK1/NCOA4-mediated inhibition of iron autophagy, and to screen for clinical targeted adjuvant therapy drugs based on this mechanism. METHODS A HCC rat model was induced with N-Nitrosodiethylamine (DEN). The physiological status of the HCC rats was assessed through indicators such as body weight and organ index. Liver damage in HCC rats was evaluated using hematoxylin and eosin (HE) staining and biochemical markers. Additionally, untargeted metabolomics was employed to explore the pathogenesis of HCC. UPLC-Q-TOF-MS combined with network pharmacology was employed to elucidate novel mechanisms, predict pathway targets, filtrate active ingredients and analyze the biological processes and signaling pathways modulated by EPME. DEN liver cancer rats were treated with different concentrations of EPME and protein expression levels were assessed by Western blot analysis. Molecular docking techniques were utilized to assess the binding affinity between the core components of EPME and target proteins. A HepG2 liver cancer in vitro model, in combination with inhibitor (SBI-0206965), was employed to verify the modulatory effects of EPME and its active ingredients on the ERK/ULK1/NCOA4 signaling pathway. Microscale thermophoretic (MST) was employed to verify the binding ability of the EPME core components to the ULK1 protein. RESULTS Metabolomics combined with network pharmacology revealed a novel pathogenesis of HCC, which is ERK/ULK1/NCOA4-mediated iron autophagy inhibition. EPME can activate iron autophagy mediated by ERK/ULK1/NCOA4 through active ingredients such as icaritin, astragalin, and emodin, thereby enhancing the survival conditions of HCC-afflicted rats and mitigating liver damage and carcinogenesis, ultimately achieving therapeutic outcomes in HCC treatment. CONCLUSION The ERK/ULK1/NCOA4-mediated iron autophagy inhibition represents a novel therapeutic mechanism for HCC. The clinical adjuvant drug EPME may exert therapeutic effects on HCC by activating ERK/ULK1/NCOA4-mediated iron autophagy.
Collapse
Affiliation(s)
- Liying Han
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China.
| | - Xiangmu Tian
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China.
| | - Xinxin Yang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China; Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang 110036, China.
| | - Tianjiao Li
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China.
| | - Shuai Wang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China; Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang 110036, China.
| | - Yongrui Bao
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China; Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang 110036, China.
| | - Xiansheng Meng
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China.
| |
Collapse
|
7
|
Li Y, Zhou F, Xu Z. TRIM25 facilitates ferroptosis in ovarian cancer through promoting PIEZO1 K63-linked ubiquitination and degradation. Transl Oncol 2025; 56:102386. [PMID: 40250035 PMCID: PMC12033990 DOI: 10.1016/j.tranon.2025.102386] [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: 01/13/2025] [Accepted: 04/05/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Ovarian cancer represents a significant threat to women's health. and ferroptosis is recognized as a potential natural inhibitor in cancer therapy, the regulatory mechanism of TRIM25 in ovarian cancer and its potential for regulating ferroptosis as a treatment remain unclear. METHODS The role of TRIM25 in ovarian cancer was examined through functional gain- and loss-of-function assays both in vitro and in vivo, while its target genes were identified. The stability and ubiquitination sites of PIEZO1 were analyzed using protein docking and ubiquitination experiments. RESULTS TRIM25 is highly expressed in ovarian cancer and promotes the growth and metastasis of ovarian cancer cells both in vivo and in vitro. Mechanistically, it facilitates PIEZO1 degradation through ubiquitination-dependent proteasome activity, inhibits ferroptosis, and stimulates ovarian cancer cell growth. CONCLUSION Our study clearly shows that TRIM25 stimulates ovarian cancer by inducing K63-linked ubiquitination of PIEZO1, which suppresses ferroptosis and promotes excessive proliferation of ovarian cancer cells. Further research identified the ubiquitination modification site on PIEZO1, providing insights for ovarian cancer treatment.
Collapse
Affiliation(s)
- Ya Li
- Department of Gynecology, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, No.12 Yancheng Road, Hengyang city, Hunan province, 421000, PR China
| | - Fei Zhou
- Department of Gynecology, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, No.12 Yancheng Road, Hengyang city, Hunan province, 421000, PR China
| | - Zhengmei Xu
- Department of Gynecology, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, No.12 Yancheng Road, Hengyang city, Hunan province, 421000, PR China.
| |
Collapse
|
8
|
Catapano A, Cimmino F, Petrella L, Pizzella A, D'Angelo M, Ambrosio K, Marino F, Sabbatini A, Petrelli M, Paolini B, Lucchin L, Cavaliere G, Cristino L, Crispino M, Trinchese G, Mollica MP. Iron metabolism and ferroptosis in health and diseases: The crucial role of mitochondria in metabolically active tissues. J Nutr Biochem 2025; 140:109888. [PMID: 40057002 DOI: 10.1016/j.jnutbio.2025.109888] [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: 05/17/2024] [Revised: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/30/2025]
Abstract
Iron is essential in various physiological processes, but its accumulation leads to oxidative stress and cell damage, thus iron homeostasis has to be tightly regulated. Ferroptosis is an iron-dependent non-apoptotic regulated cell death characterized by iron overload and reactive oxygen species accumulation. Mitochondria are organelles playing a crucial role in iron metabolism and involved in ferroptosis. MitoNEET, a protein of mitochondrial outer membrane, is a key element in this process. Ferroptosis, altering iron levels in several metabolically active organs, is linked to several non-communicable diseases. For example, iron overload in the liver leads to hepatic fibrosis and cirrhosis, accelerating non-alcholic fatty liver diseases progression, in the muscle cells contributes to oxidative damage leading to sarcopenia, and in the brain is associated to neurodegeneration. The aim of this review is to investigate the intricate balance of iron regulation focusing on the role of mitochondria and oxidative stress, and analyzing the ferroptosis implications in health and disease.
Collapse
Affiliation(s)
- Angela Catapano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy; Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Lidia Petrella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Katia Ambrosio
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Francesca Marino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Annarita Sabbatini
- Dietetic and Clinical Nutrition Unit, IEO European Institute of Oncology IRCSS, Milan, Italy
| | - Massimiliano Petrelli
- Department of Clinical and Molecular Sciences, Clinic of Endocrinology and Metabolic Diseases, Università Politecnica delle Marche, Ancona, Italy
| | - Barbara Paolini
- Department of Innovation, experimentation and clinical research, Unit of dietetics and clinical nutrition, S. Maria Alle Scotte Hospital, University of Siena, Siena, Italy
| | - Lucio Lucchin
- Dietetics and Clinical Nutrition, Bolzano Health District, Bolzano, Italy
| | - Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | | | | |
Collapse
|
9
|
Leak L, Wang Z, Joseph AJ, Johnson B, Chan AA, Decosto CM, Magtanong L, Ko PJ, Lee WC, Ritho J, Manukian S, Millner A, Chitkara S, Salinas JJ, Skouta R, Rees MG, Ronan MM, Roth JA, Myers CL, Moffat J, Boone C, Bensinger SJ, Nathanson DA, Atilla-Gokcumen GE, Moding EJ, Dixon SJ. Tegavivint triggers TECR-dependent nonapoptotic cancer cell death. Nat Chem Biol 2025:10.1038/s41589-025-01913-4. [PMID: 40419770 DOI: 10.1038/s41589-025-01913-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/16/2025] [Indexed: 05/28/2025]
Abstract
Small molecules that induce nonapoptotic cell death are of fundamental mechanistic interest and may be useful to treat certain cancers. Here we report that tegavivint, a drug candidate undergoing human clinical trials, can activate a unique mechanism of nonapoptotic cell death in sarcomas and other cancer cells. This lethal mechanism is distinct from ferroptosis, necroptosis and pyroptosis and requires the lipid metabolic enzyme trans-2,3-enoyl-CoA reductase (TECR). TECR is canonically involved in the synthesis of very-long-chain fatty acids but appears to promote nonapoptotic cell death in response to CIL56 and tegavivint via the synthesis of the saturated long-chain fatty acid palmitate. These findings outline a lipid-dependent nonapoptotic cell death mechanism that can be induced by a drug candidate currently being tested in humans.
Collapse
Affiliation(s)
- Logan Leak
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Ziwei Wang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alby J Joseph
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Brianna Johnson
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Alyssa A Chan
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | | | - Pin-Joe Ko
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Joan Ritho
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Sophia Manukian
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Alec Millner
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Shweta Chitkara
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Jennifer J Salinas
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rachid Skouta
- Department of Chemistry, University of Massachusetts, Amherst, Amherst, MA, USA
- Department of Biology, University of Massachusetts, Amherst, Amherst, MA, USA
| | | | | | | | - Chad L Myers
- Department of Computer Science and Engineering, Bioinformatics and Computational Biology Graduate Program, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Jason Moffat
- Program in Genetics & Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Steven J Bensinger
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- UCLA Lipidomics Laboratory, University of California, Los Angeles, Los Angeles, CA, USA
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - G Ekin Atilla-Gokcumen
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
10
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
11
|
Dupont S. Mechano-metabolism on the rise. Curr Opin Cell Biol 2025; 95:102529. [PMID: 40381432 DOI: 10.1016/j.ceb.2025.102529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/16/2025] [Accepted: 04/20/2025] [Indexed: 05/20/2025]
Abstract
Cells respond to the physical and geometrical tissue properties by multiple mechanotransduction mechanisms that can profoundly influence cells' decision-making, extending to cell metabolism. This review incorporates the most recent findings on this topic, organized along the idea that the mechano-metabolic connection serves three main functions, namely to inform systemic metabolism on the general functioning of a tissue/organ, to tune cells' energy production with the mechanical requirements imposed by their surroundings, and to coordinate cell metabolism with cell fate choices induced in response to mechanical cues. This connection highlights the pervasive influence of mechanical cues on cell activity, opens interesting questions on its physiological and pathological roles, and lays the foundations for exploiting the mechano-metabolism axis to design new therapeutic approaches.
Collapse
Affiliation(s)
- Sirio Dupont
- Department of Molecular Medicine, University of Padova, via Bassi 58/B, 35131, Padova, IT, Italy.
| |
Collapse
|
12
|
Chan KY, Yu Y, Kong Y, Cheng L, Yao R, Yin Chair PS, Wang P, Wang R, Sun WY, He RR, Min J, Wang F, Björklund M. GPX4-dependent ferroptosis sensitivity is a fitness trade-off for cell enlargement. iScience 2025; 28:112363. [PMID: 40330887 PMCID: PMC12053632 DOI: 10.1016/j.isci.2025.112363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/20/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Despite wide variation, each cell type has an optimal size. Maintaining optimal size is essential for cellular fitness and function but the biological basis for this remains elusive. Here, we performed fitness analysis involving genome-wide CRISPR-Cas9 knockout data from tens of human cell lines and identified that cell size influences the essentiality of genes related to mitochondria and membrane repair. These genes also included glutathione peroxidase 4 (GPX4), which safeguards membranes from oxidative damage and prevents ferroptosis-iron-dependent death. Growth beyond normal size, with or without cell-cycle arrest, increased lipid peroxidation, resulting in a ferroptosis-sensitive state. Proteomic analysis revealed cell-cycle-independent superscaling of endoplasmic reticulum, accumulation of iron, and lipidome remodeling. Even slight increases from normal cell size sensitized proliferating cells to ferroptosis as evidenced by deep-learning-based single-cell analysis. Thus, lipid peroxidation may be a fitness trade-off that constrains cell enlargement and contributes to the establishment of an optimal cell size.
Collapse
Affiliation(s)
- Kuan Yoow Chan
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Yini Yu
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Yidi Kong
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Ling Cheng
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Renzhi Yao
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Phoebe Sha Yin Chair
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Ping Wang
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Rong Wang
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Wan-Yang Sun
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Rong-Rong He
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mikael Björklund
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9JZ, UK
| |
Collapse
|
13
|
Chen Z, Xie J, Ma C, Zhang P, Lei X. Oxidative Damage Under Microgravity Conditions: Response Mechanisms, Monitoring Methods and Countermeasures on Somatic and Germ Cells. Int J Mol Sci 2025; 26:4583. [PMID: 40429729 PMCID: PMC12111329 DOI: 10.3390/ijms26104583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
With the growing human interest in space exploration, understanding the oxidative damage effects of microgravity on somatic and germ cells and their underlying mechanisms has become a pivotal scientific challenge for ensuring reproductive health during long-term space missions. In this review, we comprehensively summarize the molecular mechanisms of microgravity-induced oxidative stress, advanced detection methods, and potential protective strategies for germ cells. The evidence demonstrates that microgravity substantially compromises germ cell viability and embryonic developmental potential by disrupting mitochondrial function, increasing reactive oxygen species (ROS) production, and impairing antioxidant defenses. These alterations result in DNA damage, lipid peroxidation, and protein oxidation, thereby affecting cellular integrity and functionality. Furthermore, we discuss how cells respond to microgravity-induced oxidative stress through adaptive mechanisms, such as autophagy, apoptosis, and antioxidant systems, although these responses can have both beneficial and detrimental effects on cellular homeostasis. Additionally, this paper highlights the utility of fluorescent probes for detecting ROS levels under microgravity conditions, which are convenient and practical, but may require further optimization to improve sensitivity and specificity. To counteract these challenges, interventions such as antioxidants and artificial gravity systems show promise but need rigorous validation in prolonged microgravity environments. Finally, future research should integrate multi-omics approaches to unravel the oxidative damage network, advance space-adapted reproductive technologies, and provide essential theoretical insights and technical support for maintaining human reproductive health beyond Earth.
Collapse
Affiliation(s)
- Zekai Chen
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China;
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (J.X.); (C.M.)
| | - Jingtong Xie
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (J.X.); (C.M.)
| | - Chiyuan Ma
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (J.X.); (C.M.)
| | - Pengfei Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
| | - Xiaohua Lei
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China;
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (J.X.); (C.M.)
| |
Collapse
|
14
|
Yang JY, Luo CH, Wang KB, Tu XY, Xiao YY, Ou YT, Xie YX, Guan CX, Zhong WJ. Unraveling the mechanisms of NINJ1-mediated plasma membrane rupture in lytic cell death and related diseases. Int J Biol Macromol 2025; 309:143165. [PMID: 40239793 DOI: 10.1016/j.ijbiomac.2025.143165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/03/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
Plasma membrane rupture (PMR), the ultimate event during lytic cell death, releases damage-associated molecular patterns (DAMPs) that trigger inflammation and immune responses in the development of various diseases. Recent years have witnessed significant advances in understanding the PMR mediated by ninjurin1 (NINJ1) in different lytic cell death processes. NINJ1 oligomerizes and ruptures the membrane in pyroptosis and other lytic cell death, participating in the pathogenesis of multiple diseases. Although the membrane-permeabilizing function of NINJ1 is well recognized, the role of NINJ1 in different types of lytic cell death and its impact on multiple disease processes have yet to be fully elucidated. This review summarizes the latest advances in the mechanisms of NINJ1-mediated PMR, discusses the membrane-inducing activity of NINJ1 in different lytic cell death, explains the implications of NINJ1 in lytic cell death-related diseases, and lists the inhibitory strategies for NINJ1. We expect to provide new insights into targeting NINJ1 to suppress lytic cell death for therapeutic benefit, which may become a new strategy to control inflammatory cell lysis-related diseases.
Collapse
Affiliation(s)
- Ji-Yan Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Chen-Hua Luo
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Kun-Bo Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Xin-Yu Tu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Yun-Ying Xiao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Ye-Tong Ou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Yan-Xin Xie
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China.
| |
Collapse
|
15
|
Li C, Hu G, Huang J, Lu C, Zhang K, Shen Y, Zou Y, Zhang L, Zheng X, Chang Y. Skin mucus proteomic provides insights into the alkaline tolerance of grass carp (Ctenopharyngodon idella). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101523. [PMID: 40327979 DOI: 10.1016/j.cbd.2025.101523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
Fish skin mucus is a dynamic mucosal layer located between the epidermis and the environment, serving as the first line of defense against external environmental stresses. Studying changes in fish skin mucus under high alkaline conditions can help identify potential biomarkers of alkaline tolerance, thereby providing a basis for developing non-invasive detection methods. In this study, grass carp (Ctenopharyngodon idella) were subjected to a 7-day stress test at three different alkalinity levels: low alkalinity (10 mmol/L sodium bicarbonate), medium alkalinity (30 mmol/L sodium bicarbonate), and high alkalinity (50 mmol/L sodium bicarbonate). Fish skin mucus was collected and analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) in data-independent acquisition (DIA) mode. A total of 9656 proteins were identified across 12 samples from four groups. The low and medium alkalinity groups shared more common functional enrichments, while the high alkalinity group showed significant enrichments in pathways related to oxygen binding, the immune system, and myocarditis. More nodes and pairwise interactions were founded in the high alkalinity group. Multiple proteins related to osmoregulation, including Prostaglandin G/H Synthase 1 (PTGS1) and Group XV Phospholipase A2 (PLA2G15), are highly expressed in the high-alkalinity treatment group. These results suggest that high-alkalinity stress-induced changes in the skin mucus of grass carp reflect the continuous activation of osmoregulatory mechanisms, and the high expression of PTGS1 and PLA2G15 in the skin mucus may serve as signatures of the survival status of grass carp in alkaline conditions.
Collapse
Affiliation(s)
- Chenghao Li
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Guo Hu
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Jing Huang
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Cuiyun Lu
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Kexin Zhang
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Yubang Shen
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Yuting Zou
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Limin Zhang
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Xianhu Zheng
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Yumei Chang
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China.
| |
Collapse
|
16
|
Qiu Y, Hüther JA, Wank B, Rath A, Tykwe R, Aldrovandi M, Henkelmann B, Mergner J, Nakamura T, Laschat S, Conrad M, Stöhr D, Rehm M. Interplay of ferroptotic and apoptotic cell death and its modulation by BH3-mimetics. Cell Death Differ 2025:10.1038/s41418-025-01514-7. [PMID: 40301648 DOI: 10.1038/s41418-025-01514-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025] Open
Abstract
Ferroptosis and apoptosis are widely considered to be independent cell death modalities. Ferroptotic cell death is a consequence of insufficient radical detoxification and progressive lipid peroxidation, which is counteracted by glutathione peroxidase-4 (GPX4). Apoptotic cell death can be triggered by a wide variety of stresses, including oxygen radicals, and can be suppressed by anti-apoptotic members of the BCL-2 protein family. Mitochondria are the main interaction site of BCL-2 family members and likewise a major source of oxygen radical stress. We therefore studied if ferroptosis and apoptosis might intersect and possibly interfere with one another. Indeed, cells dying from impaired GPX4 activity displayed hallmarks of both ferroptotic and apoptotic cell death, with the latter including (transient) membrane blebbing, submaximal cytochrome-c release and caspase activation. Targeting BCL-2, MCL-1 or BCL-XL with BH3-mimetics under conditions of moderate ferroptotic stress in many cases synergistically enhanced overall cell death and frequently skewed primarily ferroptotic into apoptotic outcomes. Surprisingly though, in other cases BH3-mimetics, most notably the BCL-XL inhibitor WEHI-539, counter-intuitively suppressed cell death and promoted cell survival following GPX4 inhibition. Further studies revealed that most BH3-mimetics possess previously undescribed antioxidant activities that counteract ferroptotic cell death at commonly employed concentration ranges. Our results therefore show that ferroptosis and apoptosis can intersect. We also show that combining ferroptotic stress with BH3-mimetics, context-dependently can either enhance and convert cell death outcomes between ferroptosis and apoptosis or can also suppress cell death by intrinsic antioxidant activities.
Collapse
Affiliation(s)
- Yun Qiu
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Juliana A Hüther
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Bianca Wank
- Institute of Organic Chemistry, University of Stuttgart, Stuttgart, Germany
| | - Antonia Rath
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - René Tykwe
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Maceler Aldrovandi
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Bernhard Henkelmann
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at MRI, TUM, Munich, Germany
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Sabine Laschat
- Institute of Organic Chemistry, University of Stuttgart, Stuttgart, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Daniela Stöhr
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
17
|
Song L, Gao F, Man J. Ferroptosis: the potential key roles in idiopathic pulmonary fibrosis. Eur J Med Res 2025; 30:341. [PMID: 40296070 PMCID: PMC12036158 DOI: 10.1186/s40001-025-02623-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by recurrent injury to alveolar epithelial cells, epithelial-mesenchymal transition, and fibroblast activation, which leads to excessive deposition of extracellular matrix (ECM) proteins. However, effective preventative and therapeutic interventions are currently lacking. Ferroptosis, a unique form of iron-dependent lipid peroxidation-induced cell death, exhibits distinct morphological, physiological, and biochemical features compared to traditional programmed cell death. Recent studies have revealed a close relationship between iron homeostasis and the pathogenesis of pulmonary interstitial fibrosis. Ferroptosis exacerbates tissue damage and plays a crucial role in regulating tissue repair and the pathological processes involved. It leads to recurrent epithelial injury, where dysregulated epithelial cells undergo epithelial-mesenchymal transition via multiple signaling pathways, resulting in the excessive release of cytokines and growth factors. This dysregulated environment promotes the activation of pulmonary fibroblasts, ultimately culminating in pulmonary fibrosis. This review summarizes the latest advancements in ferroptosis research and its role in the pathogenesis and treatment of IPF, highlighting the significant potential of targeting ferroptosis for IPF management. Importantly, despite the rapid developments in this emerging research field, ferroptosis studies continue to face several challenges and issues. This review also aims to propose solutions to these challenges and discusses key concepts and pressing questions for the future exploration of ferroptosis.
Collapse
Affiliation(s)
- Longfei Song
- Department of Rehabilitation Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428 Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China
| | - Fusheng Gao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428, Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China
| | - Jun Man
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428, Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China.
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, No. 4948, Shengli East Street, Kuiwen District, Weifang City, 261041, Shandong Province, China.
| |
Collapse
|
18
|
Abdukarimov N, Kokabi K, Kunz J. Ferroptosis and Iron Homeostasis: Molecular Mechanisms and Neurodegenerative Disease Implications. Antioxidants (Basel) 2025; 14:527. [PMID: 40427409 PMCID: PMC12108473 DOI: 10.3390/antiox14050527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Iron dysregulation has emerged as a pivotal factor in neurodegenerative pathologies, especially through its capacity to promote ferroptosis, a unique form of regulated cell death driven by iron-catalyzed lipid peroxidation. This review synthesizes current evidence on the molecular underpinnings of ferroptosis, focusing on how disruptions in iron homeostasis interact with key antioxidant defenses, such as the system Xc--glutathione-GPX4 axis, to tip neurons toward lethal oxidative damage. Building on these mechanistic foundations, we explore how ferroptosis intersects with hallmark pathologies in Alzheimer's disease (AD) and Parkinson's disease (PD) and examine how iron accumulation in vulnerable brain regions may fuel disease-specific protein aggregation and neurodegeneration. We further surveyed the distinct components of ferroptosis, highlighting the role of lipid peroxidation enzymes, mitochondrial dysfunction, and recently discovered parallel pathways that either exacerbate or mitigate neuronal death. Finally, we discuss how these insights open new avenues for neuroprotective strategies, including iron chelation and lipid peroxidation inhibitors. By highlighting open questions, this review seeks to clarify the current state of knowledge and proposes directions to harness ferroptosis modulation for disease intervention.
Collapse
Affiliation(s)
| | | | - Jeannette Kunz
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan; (N.A.); (K.K.)
| |
Collapse
|
19
|
Lei J, Chen W, Gu Y, Lv X, Kang X, Jiang X. Ferroptosis regulation by traditional chinese medicine for ischemic stroke intervention based on network pharmacology and data mining. PLoS One 2025; 20:e0321751. [PMID: 40238820 PMCID: PMC12002469 DOI: 10.1371/journal.pone.0321751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/27/2025] [Indexed: 04/18/2025] Open
Abstract
OBJECTIVE The aim of this study is to use network pharmacology and data mining to explore the role of traditional Chinese medicine (TCM) in ischemic stroke (IS) intervention by ferroptosis regulation. The results will provide reference for related research on ferroptosis in IS. METHODS The ferroptosis-related targets were obtained from the GeneCards, GeneCLiP3, and FerrDdb databases, while the IS targets were sourced from the GeneCards and DisGeNET databases. Venny was used to identify IS targets associated with ferroptosis. A protein-protein interaction (PPI) analysis was then conducted, and machine learning screening was used to validate these potential targets. The potential targets that met specific criteria and their related compounds allowed us to select TCMs. A mechanistic analysis of the potential targets was conducted using the DAVID database. PPI network diagrams, target-compound network diagrams, and target-compound-TCM network diagrams were then constructed. Finally, molecular docking technology was used to verify the binding activities of the TCM compounds and core components with the identified targets. In addition, the properties, flavors, meridian tropism, and therapeutic effects of the candidate TCMs were analyzed and statistically evaluated. RESULTS A total of 706 targets associated with ferroptosis in IS were obtained, and 14 potential ferroptosis targets in IS were obtained using machine learning. Furthermore, 413 compounds and 301 TCMs were screened, and the binding activities of the targets to the TCM compounds and the core prescriptions were stable. The candidate TCMs primarily exhibited cold, warm, bitter taste, pungent taste, liver meridian, heat-cleaning medicinal, and tonify deficiency properties. CONCLUSIONS This study investigated ferroptosis regulation for IS intervention using TCM. We began by investigating the targets of IS and ferroptosis, and we also analyzed the relevant mechanism of ferroptosis in IS. The results of this study provide reference for related research on ferroptosis in IS.
Collapse
Affiliation(s)
- Jun Lei
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yaodong Gu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xueyan Lv
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xingyu Kang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xicheng Jiang
- Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
20
|
Mishima E, Nakamura T, Doll S, Proneth B, Fedorova M, Pratt DA, Friedmann Angeli JP, Dixon SJ, Wahida A, Conrad M. Recommendations for robust and reproducible research on ferroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00843-2. [PMID: 40204928 DOI: 10.1038/s41580-025-00843-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
Ferroptosis is a necrotic, non-apoptotic cell death modality triggered by unrestrained iron-dependent lipid peroxidation. By unveiling the regulatory mechanisms of ferroptosis and its relevance to various diseases, research over the past decade has positioned ferroptosis as a promising therapeutic target. The rapid growth of this research field presents challenges, associated with potentially inadequate experimental approaches that may lead to misinterpretations in the assessment of ferroptosis. Typical examples include assessing whether an observed phenotype is indeed linked to ferroptosis, and selecting appropriate animal models and small-molecule modulators of ferroptotic cell death. This Expert Recommendation outlines state-of-the-art methods and tools to reliably study ferroptosis and increase the reproducibility and robustness of experimental results. We present highly validated compounds and animal models, and discuss their advantages and limitations. Furthermore, we provide an overview of the regulatory mechanisms and the best-studied players in ferroptosis regulation, such as GPX4, FSP1, SLC7A11 and ACSL4, discussing frequent pitfalls in experimental design and relevant guidance. These recommendations are intended for researchers at all levels, including those entering the expanding and exciting field of ferroptosis research.
Collapse
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Sebastian Doll
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany.
- Translational Redox Biology, TUM Natural School of Sciences, Technical University of Munich, Garching, Germany.
| |
Collapse
|
21
|
Lee WC, Dixon SJ. Mechanisms of ferroptosis sensitization and resistance. Dev Cell 2025; 60:982-993. [PMID: 40199240 DOI: 10.1016/j.devcel.2025.02.004] [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: 09/06/2024] [Revised: 12/08/2024] [Accepted: 02/05/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis is an iron-dependent and oxidative form of non-apoptotic cell death with roles in development, homeostasis, and disease. Ferroptosis sensitivity can vary between cells, often for reasons that are not well understood. In this perspective, we describe the core ferroptosis mechanism and outline how changes in iron, redox, and lipid metabolism can alter ferroptosis sensitivity. We propose the concept of a ferroptosis sensitivity-resistance continuum to describe how different intrinsic and extrinsic factors interact to push cells toward a more ferroptosis-sensitive or ferroptosis-resistant state, with effects on development and diseases such as cancer.
Collapse
Affiliation(s)
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Liu B, Yao X, Huang Q, Shi Z, Wei J, Li S, Li M, Chen X, Dai J. Sodium fluoride promotes myopia progression via the activation of the ferroptosis pathway by PIEZO1 and pharmacological targeting PIEZO1 represents an innovative approach for myopia treatment. Cell Biol Toxicol 2025; 41:64. [PMID: 40175653 PMCID: PMC11965261 DOI: 10.1007/s10565-025-10020-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025]
Abstract
Sodium fluoride-induced ocular damage constitutes a significant public health concern globally; however, the precise molecular mechanisms underlying this issue remain obscure. This study aims to investigate the effects of sodium fluoride on myopia and to offer novel theoretical foundations for future strategies in myopia prevention and control. The experimental data showed that sodium fluoride could promote myopia progression, and through bioinformatics analysis, we found that sodium fluoride could affect the ferroptosis pathway. Western blotting and redox kit assays further confirmed that sodium fluoride activates the ferroptosis pathway. We also demonstrated that PIEZO1 plays a crucial role in sodium fluoride-induced myopia, and that the PIEZO1 inhibitor (GsMTx4) can inhibit the ferroptosis pathway. Subsequently, we identified PIEZO1 as a potential target of baicalin, which inhibited PIEZO1 expression in vivo and in vitro, as confirmed by molecular docking modeling and CETSA assays. Finally, we found that baicalin inhibited sodium fluoride-induced myopia via PIEZO1. Taken together, our findings indicate that sodium fluoride can promote myopia progression by activating the ferroptosis pathway through PIEZO1, and that targeting PIEZO1 expression can delay myopia progression, which may provide a new drug target for myopia treatment in the future.
Collapse
Affiliation(s)
- Bo Liu
- Department of Ophthalmology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Xueting Yao
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qinying Huang
- Department of Ophthalmology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zehui Shi
- Department of Ophthalmology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Jinfei Wei
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shijia Li
- Shanghai Sixth People'S Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Li
- Department of Ophthalmology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xiuping Chen
- Department of Ophthalmology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China.
| | - Jinhui Dai
- Department of Ophthalmology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China.
| |
Collapse
|
23
|
Lancaster GI, Murphy AJ. Do physiological changes in fatty acid composition alter cellular ferroptosis susceptibility and influence cell function? J Lipid Res 2025; 66:100765. [PMID: 40021010 PMCID: PMC11981815 DOI: 10.1016/j.jlr.2025.100765] [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: 12/19/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Ferroptosis is an iron-dependent form of cell death driven by the excessive peroxidation of poly-unsaturated fatty acids (PUFAs) within membrane phospholipids. Ferroptosis is a hallmark of many diseases and preventing or inducing ferroptosis has considerable therapeutic potential. Like other forms of cell death, the pathological importance and therapeutic potential of ferroptosis is well appreciated. However, while cell death modalities such as apoptosis and necroptosis have critical physiological roles, such as in development and tissue homeostasis, whether ferroptosis has important physiological roles is largely unknown. In this regard, key questions for field are as follows: Is ferroptosis used for physiological processes? Are certain cell-types purposely adapted to be either resistant or sensitive to ferroptosis to be able to function optimally? Do physiological perturbations such as aging and diet impact ferroptosis susceptibility? Herein, we have reviewed emerging evidence that supports the idea that being able to selectively and controllably induce or resist ferroptosis is essential for development and cell function. While several factors regulate ferroptosis, it appears that the ability of cells and tissues to control their lipid composition, specifically the abundance of phospholipids containing PUFAs, is crucial for cells to be able to either resist or be sensitized to ferroptosis. Finally, aging and diets enriched in specific PUFAs lead to an increase in cellular PUFA levels which may sensitize cells to ferroptosis. Therefore, changes in dietary PUFAs or againg may impact the pathogenesis of diseases where ferroptosis is involved.
Collapse
Affiliation(s)
- Graeme I Lancaster
- Department of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Immunology, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| | - Andrew J Murphy
- Department of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Immunology, Monash University, Melbourne, Victoria, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
24
|
Pan L, He B, Han Y, Yuan D, Duan X, Wang Y. Yanghe Pingchuan granules induce ferroptosis in airway smooth muscle cells to improve bronchial asthma via the METTL3/P53/SLC7A11 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156480. [PMID: 39978273 DOI: 10.1016/j.phymed.2025.156480] [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: 09/03/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Recent studies have found that ferroptosis is strongly associated with the development of bronchial asthma (BA). However, the mechanism underlying the role of ferroptosis in asthma remains unclear. Yanghe Pingchuan granules (YPG) have significant curative effect in the clinical treatment of BA. In our previous study, we found that YPG inhibit pyroptosis in the airway smooth muscle cells (ASMCs) of and reducing airway inflammation. Whether ferroptosis participated in the YPG treated BA activity is an interesting project. PURPOSE The aim of this study was to investigate the protective effects and the related mechanisms of YPG against BA. METHODS We used ultra high-performance liquid chromatograph (UPLC) to analyze the composition of YPG. Ovalbumin (OVA)-induced BA models were developed in vivo. YPG was administered to rats by gavage and ASMCs were isolated and cultured using α-SMA and CCK8 was used to assess cell viability. Gene editing, m6A RNA immunoprecipitation (MeRIP), western blotting, RT-qPCR, and transmission electron microscopy (TEM) was used to assess ferroptosis protein and mRNA expression in ASMCs. Further, the mechanism of YPG-induced regulation of ferroptosis in ASMCs via the METTL3/P53/SLC7A11 signaling axis was interrogated. BA rats were used to verify the therapeutic effects and mechanism of YPG. Moreover, hematoxylin and eosin staining was used to evaluate pathological changes using animal samples, while immunofluorescence, western blotting, RT-qPCR, and TEM were used to verify the mechanism by which YPG improved BA through the METTL3/P53/SLC7A11 signaling axis. RESULTS Qualitative analysis revealed seven major components in YPG. Our in vivo and in vitro data confirm that YPG significantly induced ferroptosis in ASMCs. YPG treatment effectively increased the expression of Fe2+, P53, and PTGS2, while decreasing SLC7A11, GPX4, and FTH1 expression. Moreover, TEM data revealed that YPG-induced mitochondrial membrane rupture and ridge disappearance. Additionally, YPG significantly increased METTL3 expression levels and upregulated the levels of P53 m6A, thus promoting its degradation. Notably, overexpression of METTL3 and P53 induces ferroptosis of ASMCs BA rats. CONCLUSION We show that YPG may induce ferroptosis of ASMCs in BA rats by activating the METTL3/P53/SLC7A11 signaling pathway, thus alleviating disease symptoms.
Collapse
Affiliation(s)
- Lingyu Pan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Bangfu He
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China; Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yanquan Han
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Dezhi Yuan
- Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xianchun Duan
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yongzhong Wang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China.
| |
Collapse
|
25
|
Kunishige R, Noguchi Y, Okamoto N, Li L, Ono A, Murata M, Kano F. Protein covariation networks for elucidating ferroptosis inducer mechanisms and potential synergistic drug targets. Commun Biol 2025; 8:480. [PMID: 40164758 PMCID: PMC11958834 DOI: 10.1038/s42003-025-07886-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
In drug development, systematically characterizing a compound's mechanism of action (MoA), including its direct targets and effector proteins, is crucial yet challenging. Network-based approaches, unlike those focused solely on direct targets, effectively detect a wide range of cellular responses elicited by compounds. This study applied protein covariation network analysis, leveraging quantitative, morphological, and localization features from immunostained microscopic images, to elucidate the MoA of AX-53802, a novel ferroptosis inducer. From the candidate targets extracted through network analysis, GPX4 was verified as the direct target by validation experiments. Additionally, aggregates involving GPX4, TfR1, and F-actin were observed alongside iron reduction, suggesting a ferroptosis defense mechanism. Furthermore, combination therapies targeting GPX4 and FAK/Src were found to enhance cancer cell death, and MDM2, ezrin, and cortactin were identified as potential ferroptosis inhibitor targets. These findings highlight the effectiveness of network-based approaches in uncovering a compound's MoA and developing combination therapies for cancer.
Collapse
Affiliation(s)
- Rina Kunishige
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Yoshiyuki Noguchi
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
- International Research Center for Neurointelligence, Institutes for Advanced Study, The University of Tokyo, Tokyo, Japan
| | | | - Lei Li
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Akito Ono
- Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Masayuki Murata
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Fumi Kano
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan.
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan.
- Cell Biology Center, Institute of Integrated Research, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan.
| |
Collapse
|
26
|
Ben Zichri- David S, Shkuri L, Ast T. Pulling back the mitochondria's iron curtain. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:6. [PMID: 40052109 PMCID: PMC11879881 DOI: 10.1038/s44324-024-00045-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/09/2024] [Indexed: 03/09/2025]
Abstract
Mitochondrial functionality and cellular iron homeostasis are closely intertwined. Mitochondria are biosynthetic hubs for essential iron cofactors such as iron-sulfur (Fe-S) clusters and heme. These cofactors, in turn, enable key mitochondrial pathways, such as energy and metabolite production. Mishandling of mitochondrial iron is associated with a spectrum of human pathologies ranging from rare genetic disorders to common conditions. Here, we review mitochondrial iron utilization and its intersection with disease.
Collapse
Affiliation(s)
| | - Liraz Shkuri
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001 Israel
| | - Tslil Ast
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001 Israel
| |
Collapse
|
27
|
Zhang R, Sougawa N, Mao D, Inoue H, Goda S. Signaling pathways of pro-IL-1β production induced by mechanical stress in gingival epithelial cells. J Oral Biosci 2025; 67:100626. [PMID: 39921162 DOI: 10.1016/j.job.2025.100626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
OBJECTIVES Mechanical stress on the teeth and alveolar bone caused by bruxism, orthodontics, and implants affects the periodontal tissues, causing gingival recession and alveolar bone resorption, and entire body, including the heart and vascular system. Although the same forces exerted on the alveolar bone and teeth are exerted on gingival epithelial cells, little is known about the effects of mechanical stress on these cells. This study investigated the effects of mechanical stress on gingival epithelial cells. METHODS Ca9-22 cells (human gingival epithelial cells) were used. They were seeded onto the silicone and stretched cyclically. Mechanical stress-stimulated Ca9-22 cells were evaluated for pro-inflammatory interleukin (pro-IL)-1β production using Western blotting and analyzed to assess the phosphorylation level of intracellular signaling molecules. RESULTS Mechanical stress induced pro-IL-1β upregulation in Ca9-22 cells, which was significantly inhibited by ruthenium red. Ruthenium red significantly inhibited mechanical stress-induced phosphorylation of focal adhesion kinase (FAK), P130cas, and extracellular signal-regulated kinase 1 and 2 (ERK1/2) induced by mechanical stress. Additionally, Y15 significantly inhibited the upregulation of pro-IL-1β expression and phosphorylation of FAK, P130cas, and ERK1/2 stimulated by mechanical stress. CONCLUSIONS In Ca9-22 cells, mechanical stress may increase pro-IL-1β production via mechanosensitive ion channels and FAK. These findings revealed the mechanisms of inflammation in mechanically-stressed Ca9-22 cells and may aid in the development of therapeutic approaches to prevent bone resorption.
Collapse
Affiliation(s)
- Ruixuan Zhang
- Graduate School of Dentistry, Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Nagako Sougawa
- Department of Physiology, Osaka Dental University, Osaka, Japan.
| | - Dan Mao
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Hiroshi Inoue
- Department of Physiology, Osaka Dental University, Osaka, Japan
| | - Seiji Goda
- Department of Physiology, Osaka Dental University, Osaka, Japan
| |
Collapse
|
28
|
Chen SY, Shyu IL, Chi JT. NINJ1 in Cell Death and Ferroptosis: Implications for Tumor Invasion and Metastasis. Cancers (Basel) 2025; 17:800. [PMID: 40075648 PMCID: PMC11898531 DOI: 10.3390/cancers17050800] [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: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
NINJ1 was initially recognized for its role in nerve regeneration and cellular adhesion. Subsequent studies have uncovered its participation in cancer progression, where NINJ1 regulates critical steps in tumor metastasis, such as cell migration and invasion. More recently, NINJ1 has emerged as a multifunctional protein mediating plasma membrane rupture (PMR) in several lytic cell death processes, including apoptosis, necroptosis, and pyroptosis. However, its role in ferroptosis-an iron-dependent form of lytic cell death characterized by lipid peroxidation-remained unclear until 2024. Ferroptosis is a tumor suppression mechanism that may be particularly relevant to detached and metastatic cancer cells. This review explores the role of NINJ1 in tumor invasion and metastasis, focusing on its regulation of ferroptosis via a non-canonical mechanism distinct from other cell deaths. We discuss the process of ferroptosis and its implications for cancer invasion and metastasis. Furthermore, we review recent studies highlighting the diverse roles of NINJ1 in ferroptosis regulation, including its canonical function in PMR and its non-canonical function of modulating intracellular levels of glutathione (GSH) and coenzyme A (CoA) via interaction with xCT anti-porter. Given that ferroptosis has been associated with tumor suppression, metastasis, the elimination of treatment-resistant cancer cells, and tumor dormancy, NINJ1's modulation of ferroptosis presents a promising therapeutic target for inhibiting metastasis. Understanding the dual role of NINJ1 in promoting or restraining ferroptosis depending on cellular context could open avenues for novel anti-cancer strategies to enhance ferroptotic vulnerability in metastatic tumors.
Collapse
Affiliation(s)
- Ssu-Yu Chen
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA;
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ing-Luen Shyu
- Department of Obstetrics and Gynecology, Chi Mei Medical Center, Tainan 710, Taiwan
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Center for Advanced Genomic Technologies, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
29
|
Usuda J, Yagyu K, Tanaka H, Hori M, Ishikawa K, Takahashi Y. Nanoscale visualization of the anti-tumor effect of a plasma-activated Ringer's lactate solution. Faraday Discuss 2025; 257:212-223. [PMID: 39470167 DOI: 10.1039/d4fd00116h] [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: 10/30/2024]
Abstract
Plasma-activated Ringer's lactate solutions (PALs), which are Ringer's lactate solutions treated with non-thermal atmospheric-pressure plasma, have an anti-tumor effect and can be used for chemotherapy. As the anti-tumor effect of the PAL is influenced by the cell-treatment time, it is necessary to monitor the structural changes of the cell surface with non-invasive, nanoscale, and time-lapse imaging to understand the anti-tumor effect. In this study, to characterize the anti-tumor effect of the PAL, we used scanning ion conductance microscopy (SICM), using glass nanopipettes as probes, to visualize the structural changes of the cell surface. SICM time-lapse topographic imaging visualized a decrease in the movement of lamellipodia in normal cells and cancer cells after the PAL treatment. Furthermore, in normal cells, protrusive structures were observed on the cell surface. Time-lapse imaging using SICM allowed us to characterize the differences in the morphological changes between the normal and cancer cells upon exposure to the PAL.
Collapse
Affiliation(s)
- Junichi Usuda
- Department of Electronics, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Kenshin Yagyu
- Department of Electronics, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
| | - Hiromasa Tanaka
- Center for Low-Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Masaru Hori
- Center for Low-Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Kenji Ishikawa
- Center for Low-Temperature Plasma Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Yasufumi Takahashi
- Department of Electronics, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa 920-1192, Japan
| |
Collapse
|
30
|
Xu H, Yang L, Wu Y, Lei H. Double-edged sword effect of GPX4 in skin homeostasis and diseases. Arch Dermatol Res 2025; 317:404. [PMID: 39951160 DOI: 10.1007/s00403-025-03903-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/19/2025] [Accepted: 01/27/2025] [Indexed: 05/09/2025]
Abstract
Glutathione peroxidase 4 (GPX4) is a crucial antioxidant enzyme that plays a vital role in protecting cells from oxidative damage and lipid peroxidation. In the context of skin diseases, GPX4 serves as a key regulator of oxidative stress and inflammation, both of which are significant features of various skin conditions. By preventing lipid peroxidation and maintaining membrane integrity, GPX4 acts as a safeguard against cell death pathways, particularly ferroptosis, in skin diseases. Dysregulation of GPX4 in conditions such as dermatitis, psoriasis, and skin cancer is linked to heightened oxidative stress, inflammation, and tissue damage. Understanding the role of GPX4 and its intricate interactions in skin disease pathogenesis can aid in more effectively targeting oxidative stress and inflammation, leading to promising therapeutic interventions. This review summarizes the role of GPX4 in maintaining skin homeostasis and its involvement in disease, proposing strategies to target GPX4, including its post-translational modifications. Investigate the precise mechanism through which GPX4 influences the onset of skin diseases, and utilize GPX4 agonists or inhibitors as potential treatments.
Collapse
Affiliation(s)
- Hanzhang Xu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingli Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pathophysiology, Research Unit of Stress and Cancer, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hu Lei
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
31
|
Dixon S, Leak L, Wang Z, Lee WC, Johnson B, Millner A, Ko PJ, Decosto C, Magtanong L, Ritho J, Skouta R, Atilla-Gokcumen E, Myers C, Moffat J, Boone C, Bensinger S, Moding E, Joseph A, Chan A, Chitkara S, Salinas J, Nathanson D. A clinical drug candidate that triggers non-apoptotic cancer cell death. RESEARCH SQUARE 2025:rs.3.rs-4138879. [PMID: 39989975 PMCID: PMC11844650 DOI: 10.21203/rs.3.rs-4138879/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Small molecules that induce non-apoptotic cell death are of fundamental mechanistic interest and may be useful to treat certain cancers. Here, we report that tegavivint, a drug candidate undergoing human clinical trials, can activate a unique mechanism of non-apoptotic cell death in sarcomas and other cancer cells. This lethal mechanism is distinct from ferroptosis, necroptosis and pyroptosis and requires the lipid metabolic enzyme trans-2,3-enoyl-CoA reductase (TECR). TECR is canonically involved in the synthesis of very long chain fatty acids but appears to promote non-apoptotic cell death in response to CIL56 and tegavivint via the synthesis of the saturated long-chain fatty acid palmitate. These findings outline a lipid-dependent non-apoptotic cell death mechanism that can be induced by a drug candidate currently being tested in humans.
Collapse
|
32
|
Fan S, Hu Y, Shi J. Role of ferroptosis in atrial fibrillation: a review. Front Pharmacol 2025; 16:1362060. [PMID: 39981174 PMCID: PMC11839810 DOI: 10.3389/fphar.2025.1362060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 01/13/2025] [Indexed: 02/22/2025] Open
Abstract
Cardiovascular disease remains the leading cause of mortality, with atrial fibrillation emerging as one of the most common conditions encountered in clinical practice. However, its underlying mechanisms remain poorly understood, prompting ongoing research. Ferroptosis, a recently discovered form of regulated cell death characterized by lipid peroxidation and disrupted cellular redox balance leading to cell death due to iron overload, has attracted significant attention. Since its identification, ferroptosis has been extensively studied in various contexts, including cancer, stroke, myocardial ischemia/reperfusion injury, and heart failure. Growing evidence suggests that ferroptosis may also play a critical role in the onset and progression of atrial fibrillation, though research in this area is still limited. This article provides a concise overview of the potential mechanisms by which ferroptosis may contribute to the pathogenesis of atrial fibrillation.
Collapse
Affiliation(s)
- Shaowei Fan
- Lugouqiao Second Community Health Service Center, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, China
| | - Yuanhui Hu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Jingjing Shi
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
33
|
Zhao G, Li X, Zhang Y, Wang X, Deng L, Xu J, Jin S, Zuo Z, Xun L, Luo M, Yang F, Qi J, Fu P. Intricating connections: the role of ferroptosis in systemic lupus erythematosus. Front Immunol 2025; 16:1534926. [PMID: 39967676 PMCID: PMC11832682 DOI: 10.3389/fimmu.2025.1534926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory and autoimmune disease with multiple tissue damage. However, the pathology remains elusive, and effective treatments are lacking. Multiple types of programmed cell death (PCD) implicated in SLE progression have recently been identified. Although ferroptosis, an iron-dependent form of cell death, has numerous pathophysiological features similar to those of SLE, such as intracellular iron accumulation, mitochondrial dysfunction, lipid metabolism disorders and concentration of damage associated-molecular patterns (DAMPs), only a few reports have demonstrated that ferroptosis is involved in SLE progression and that the role of ferroptosis in SLE pathogenesis continues to be neglected. Therefore, this review elucidates the potential intricate relationship between SLE and ferroptosis to provide a reliable theoretical basis for further research on ferroptosis in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Guowang Zhao
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xinghai Li
- Department of Minimal Invasive Intervention Radiology, Ganzhou People’s Hospital, Ganzhou, Jiangxi, China
| | - Ying Zhang
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xingzi Wang
- Department of Nephrology, Yueyang Central Hospital, Yueyang, Hunan, China
| | - Li Deng
- Department of Internal Medicine, Community Health Service Station of Dian Mian Avenue, Kunming, Yunnan, China
| | - Juan Xu
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Shumei Jin
- Yunnan Institute of Food and Drug Supervision and Control, Medical Products Administration of Yunnan Province, Kunming, Yunnan, China
| | - Zan Zuo
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Linting Xun
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Mei Luo
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Fan Yang
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jialong Qi
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Affiliated by Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, The First People’s Hospital of Yunnan Province, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Ping Fu
- Department of Rheumatology and Clinical Immunology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
34
|
Lu Y, Li T, Shu Y, Lu C, Luo Z, Wang J, Xiong H, Li W. Lipid peroxidation and sarcopenia: molecular mechanisms and potential therapeutic approaches. Front Med (Lausanne) 2025; 12:1525205. [PMID: 39963429 PMCID: PMC11831367 DOI: 10.3389/fmed.2025.1525205] [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: 11/09/2024] [Accepted: 01/02/2025] [Indexed: 02/20/2025] Open
Abstract
Sarcopenia is an age-related condition characterized by the progressive loss of skeletal muscle mass and strength. With the global aging population, its incidence is rapidly increasing. Lipid peroxidation is a critical biochemical process that generates reactive oxygen species (ROS), leading to the destruction of muscle cell structure and function. It plays a pivotal role in the onset and progression of sarcopenia. This review summarizes the mechanisms by which lipid peroxidation contributes to sarcopenia, with a focus on its regulatory effects on cell membrane damage, mitochondrial dysfunction, and cell death. In addition, we discuss the protective role of antioxidant factors such as GPX4 (glutathione peroxidase 4) and antioxidant peptides like SS peptides in mitigating lipid peroxidation and delaying the progression of sarcopenia. Finally, the potential of various strategies, including natural compounds, supplements, natural extracts, and lifestyle interventions, in inhibiting lipid peroxidation and promoting muscle health is explored.
Collapse
Affiliation(s)
- Yifan Lu
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Tiao Li
- Department of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Yang Shu
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chengyin Lu
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Zhiqiang Luo
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Jingrui Wang
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Hui Xiong
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Wangyang Li
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Graduate School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
35
|
Ji Y, Liu H, Niu F, Kang B, Luo X, Yang H, Tian Z, Yang J. Endoplasmic Reticulum Stress Promotes Neuronal Damage in Neonatal Hypoxic-Ischemic Brain Damage by Inducing Ferroptosis. Mol Biotechnol 2025; 67:805-815. [PMID: 38329706 DOI: 10.1007/s12033-024-01095-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
Hypoxic-ischemic brain damage (HIBD) poses a significant risk of neurological damage in newborns. This study investigates the impact of endoplasmic reticulum stress (ERS) on neuronal damage in neonatal HIBD and its underlying mechanisms. HIBD neonatal rat model was constructed and pre-treated with 4-phenylbutiric acid (4-PBA). Nissl and TUNEL staining were utilised to assess neuronal damage and apoptosis in rat brains. HIBD cell model was established by inducing oxygen-glucose deprivation (OGD) in rat H19-7 neurons, which were then pre-treated with Thapsigargin (TG), Ferrostatin-1 (Fer-1), or both. Cell viability and apoptosis of H19-7 neurons were analysed using cell counting kit-8 assay and TUNEL staining. GRP78-PERK-CHOP pathway activity and glutathione peroxidase-4 (GPX4) expression in rat brains and H19-7 neurons were assessed using Western blot. Ferroptosis-related indicators, including glutathione (GSH), superoxide dismutase (SOD), malondialdehyde (MDA) and iron content, were measured using commercial kits in both rat brains and H19-7 neurons. GRP78-PERK-CHOP pathway was overactivated in HIBD neonatal rats' brains, which was mitigated by 4-PBA treatment. 4-PBA treatment demonstrated a reduction in neuronal damage and apoptosis in HIBD-affected neonatal rat brains. Furthermore, it attenuated ferroptosis in rats by increasing GPX4, GSH and SOD while decreasing MDA and iron content. In the OGD-induced H19-7 neurons, Fer-1 treatment counteracted the suppressive effects of TG on viability, the exacerbation of apoptosis, the promotion of ferroptosis and the activation of the GRP78-PERK-CHOP pathway. Overall, ERS facilitates neuronal damage in neonatal HIBD by inducing ferroptosis. Consequently, the suppression of ERS may represent a promising therapeutic strategy for treating neonatal HIBD.
Collapse
Affiliation(s)
- Yongjia Ji
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), 127 Hupan Road, Jinfeng District, Yinchuan City, Ningxia, 750001, China.
| | - Huili Liu
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), 127 Hupan Road, Jinfeng District, Yinchuan City, Ningxia, 750001, China
| | - Fang Niu
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), 127 Hupan Road, Jinfeng District, Yinchuan City, Ningxia, 750001, China
| | - Bo Kang
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), 127 Hupan Road, Jinfeng District, Yinchuan City, Ningxia, 750001, China
| | - Xiu Luo
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), 127 Hupan Road, Jinfeng District, Yinchuan City, Ningxia, 750001, China
| | - Hua Yang
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), 127 Hupan Road, Jinfeng District, Yinchuan City, Ningxia, 750001, China
| | - Zhen Tian
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), 127 Hupan Road, Jinfeng District, Yinchuan City, Ningxia, 750001, China
| | - Juan Yang
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), 127 Hupan Road, Jinfeng District, Yinchuan City, Ningxia, 750001, China
| |
Collapse
|
36
|
Zhu L, Xu Y. Multifaceted roles of ninjurin1 in immunity, cell death, and disease. Front Immunol 2025; 16:1519519. [PMID: 39958360 PMCID: PMC11825492 DOI: 10.3389/fimmu.2025.1519519] [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: 11/14/2024] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Abstract
Ninjurin1 (NINJ1) is initially identified as a nerve injury-induced adhesion molecule that facilitates axon growth. It is initially characterized to promote nerve regeneration and mediate the transendothelial transport of monocytes/macrophages associated with neuroinflammation. Recent evidence indicates that NINJ1 mediates plasma membrane rupture (PMR) in lytic cell death. The absence or inhibition of NINJ1 can delay PMR, thereby mitigating the spread of inflammation resulting from cell lysis and preventing the progression of various cell death-related pathologies, suggesting a conserved regulatory mechanism across these processes. Further research elucidated the structural basis and mechanism of NINJ1-mediated PMR. Although the role of NINJ1 in PMR is established, the identity of its activating factors and its implications in diseases remain to be fully explored. This review synthesizes current knowledge regarding the structural basis and mechanism of NINJ1-mediated PMR and discusses its significance and therapeutic targeting potential in inflammatory diseases, neurological disorders, cancer, and vascular injuries.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, China
| | - Yunfei Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
- Postdoctoral Research Station of Biology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
37
|
Carbone GG, Mariano S, Gabriele A, Cennamo S, Primiceri V, Aziz MR, Panzarini E, Calcagnile L. Exploring the Potential of Gold Nanoparticles in Proton Therapy: Mechanisms, Advances, and Clinical Horizons. Pharmaceutics 2025; 17:176. [PMID: 40006543 PMCID: PMC11859620 DOI: 10.3390/pharmaceutics17020176] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Proton therapy represents a groundbreaking advancement in cancer radiotherapy, leveraging the unique spatial energy distribution of protons to deliver precise, high-dose radiation to tumors while sparing surrounding healthy tissues. Despite its clinical success, proton therapy faces challenges in optimizing its therapeutic precision and efficacy. Recent research has highlighted the potential of gold nanoparticles to enhance proton therapy outcomes. Due to their high atomic number and favorable biological properties, gold nanoparticles act as radiosensitizers by amplifying the generation of secondary electrons and reactive oxygen species upon proton irradiation. This enhances DNA damage in tumor cells while preserving healthy tissues. Additionally, functionalization of gold nanoparticles with tumor-targeting ligands offers improved precision, making proton therapy more effective against a broader range of cancers. This review synthesizes current knowledge on the mechanisms of gold nanoparticle radiosensitization, preclinical evidence, and the technological hurdles that must be addressed to integrate this promising approach into clinical practice, aiming to advance the efficacy and accessibility of proton therapy in cancer therapy.
Collapse
Affiliation(s)
- Giorgio Giuseppe Carbone
- CEDAD (Center of Applied Physics, Datation and Diagnostics), Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy; (G.G.C.); (A.G.); (S.C.); (V.P.); (M.R.A.); (L.C.)
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy
| | - Stefania Mariano
- CEDAD (Center of Applied Physics, Datation and Diagnostics), Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy; (G.G.C.); (A.G.); (S.C.); (V.P.); (M.R.A.); (L.C.)
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy
| | - Alessandra Gabriele
- CEDAD (Center of Applied Physics, Datation and Diagnostics), Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy; (G.G.C.); (A.G.); (S.C.); (V.P.); (M.R.A.); (L.C.)
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy
| | - Sabrina Cennamo
- CEDAD (Center of Applied Physics, Datation and Diagnostics), Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy; (G.G.C.); (A.G.); (S.C.); (V.P.); (M.R.A.); (L.C.)
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy
| | - Vitantonio Primiceri
- CEDAD (Center of Applied Physics, Datation and Diagnostics), Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy; (G.G.C.); (A.G.); (S.C.); (V.P.); (M.R.A.); (L.C.)
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy
| | - Muhammad Rizwan Aziz
- CEDAD (Center of Applied Physics, Datation and Diagnostics), Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy; (G.G.C.); (A.G.); (S.C.); (V.P.); (M.R.A.); (L.C.)
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy
| | - Elisa Panzarini
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy;
| | - Lucio Calcagnile
- CEDAD (Center of Applied Physics, Datation and Diagnostics), Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy; (G.G.C.); (A.G.); (S.C.); (V.P.); (M.R.A.); (L.C.)
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, 72100 Lecce, Italy
| |
Collapse
|
38
|
Pan Y, Matsunaga T, Zhang T, Akaike T. The Therapeutic Potential of Supersulfides in Oxidative Stress-Related Diseases. Biomolecules 2025; 15:172. [PMID: 40001475 PMCID: PMC11852411 DOI: 10.3390/biom15020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Oxidation-reduction (redox) reactions are fundamental to sustaining life, with reactive oxygen and nitrogen species playing pivotal roles in cellular signaling and homeostasis. However, excessive oxidative stress disrupts redox balance, contributing to a wide range of diseases, including inflammatory and pulmonary disorders, neurodegeneration, and cancer. Although numerous antioxidant therapies have been developed and tested for oxidative stress-related diseases, their clinical efficacy remains limited. Here, we introduce the emerging concept of 'supersulfides', a class of redox molecule species with unique antioxidant and nucleophilic properties, which have recently been recognized as crucial regulators of cellular redox homeostasis. Unlike traditional antioxidants, supersulfides offer novel mechanisms of action that directly target the underlying processes of oxidative stress. This review summarizes current knowledge on supersulfides, highlighting their roles in oxidative stress and associated diseases, as well as the mechanisms underlying oxidative stress-related pathology. The therapeutic potential of synthetic supersulfides for treating oxidative stress-related diseases is also discussed. A comprehensive understanding of the molecular and cellular basis of redox biology can help to guide the development of innovative redox-based therapeutic strategies aimed at preventing and treating diseases associated with disturbed redox regulation.
Collapse
Grants
- 20348438 Japan Science and Technology Agency
- 21H05263 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 21H05258 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 23K20040 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 24H00063 Ministry of Education, Culture, Sports, Science and Technology of Japan
- JP21zf0127001 Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Yuexuan Pan
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan;
| | - Tetsuro Matsunaga
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita 010-8543, Japan;
| | - Tianli Zhang
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita 010-8543, Japan;
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan;
| |
Collapse
|
39
|
Yuan Q, Wang M, Zhang Z, Wang R, Wang D, Sang Z, Zhao P, Liu X, Zhu X, Liang G, Fan H, Wang D. The ameliorative effects of melatonin against BDE-47-induced hippocampal neuronal ferroptosis and cognitive dysfunction through Nrf2-Chaperone-mediated autophagy of ACSL4 degradation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117542. [PMID: 39700775 DOI: 10.1016/j.ecoenv.2024.117542] [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: 07/29/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Recent studies demonstrate that lipid peroxidation-induced ferroptosis participates in 2,2',4,4'-tetrabromodiphenyl ether (BDE-47)-evoked neurotoxicity and cognitive dysfunction. Melatonin has been indicated to confer neuroprotection against brain diseases via its potent anti-ferroptotic effects. Therefore, this study aims to explore whether melatonin can mitigate BDE-47-elicited cognitive impairment via suppressing ferroptosis, and further delineate the underlying mechanisms. Our results found that melatonin administration effectively inhibited BDE-47-induced ferroptosis in mice hippocampi and murine hippocampal neuronal HT-22 cells. Acyl-CoA synthetase long-chain family member 4 (ACSL4), a key lipid metabolism enzyme dictating ferroptosis sensitivity, accompanied by higher MDA and lipid reactive oxygen species (ROS), was remarkably increased under BDE-47 stress, while melatonin supplementation could suppress the elevated ACSL4 in vivo and in vitro. Furthermore, melatonin facilitated lysosomal ACSL4 degradation through enhancing lysosome-associated membrane protein type 2a (LAMP2a) expression and chaperone-mediated autophagy (CMA) activity, while LAMP2a knockdown abrogated the positive effects of melatonin on ACSL4 elimination in BDE-47-treated HT-22 cells. Moreover, nuclear factor erythroid 2-related factor 2 (Nrf2) activation by melatonin contributed to LAMP2a upregulation and CMA of ACSL4 and subsequent neuronal ferroptosis. Importantly, melatonin, CMA activator CA77.1, and ACSL4 inhibitor rosiglitazone (RSG) administration substantially attenuated neuronal/synaptic injury and cognitive deficits following BDE-47 exposure. Taken together, these findings revealed that melatonin could prevent BDE-47-provoked ferroptosis in the hippocampal neurons and mitigate cognitive dysfunction by facilitating ACSL4 degradation via Nrf2-chaperone-mediated autophagy. Therefore, melatonin might be a potential candidate for treating BDE-47-elicited neurotoxicity and neurobehavioral disorder.
Collapse
Affiliation(s)
- Quan Yuan
- Henan Province Rongkang Hospital, Luoyang, China
| | - Mingwei Wang
- The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Zhaoxiang Zhang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Ruofei Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Dechao Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Zichun Sang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Pu Zhao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoli Liu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiaoying Zhu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Gaofeng Liang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China.
| | - Dongmei Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
40
|
Gao K, Lv L, Li Z, Wang C, Zhang J, Qiu D, Xue H, Xu Z, Tan G. Natural Products in the Prevention of Degenerative Bone and Joint Diseases: Mechanisms Based on the Regulation of Ferroptosis. Phytother Res 2025; 39:162-188. [PMID: 39513459 DOI: 10.1002/ptr.8366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 11/15/2024]
Abstract
Degenerative bone and joint diseases (DBJDs), characterized by osteoporosis, osteoarthritis, and chronic inflammation of surrounding soft tissues, are systemic conditions primarily affecting the skeletal system. Ferroptosis, a programmed cell death pathway distinct from apoptosis, autophagy, and necroptosis. Accumulating evidence suggests that ferroptosis is intricately linked to the pathogenesis of DBJDs, and targeting its regulation could be beneficial in managing these conditions. Natural products, known for their anti-inflammatory and antioxidant properties, have shown unique advantages in preventing DBJDs, potentially through modulating ferroptosis. This article provides an overview of the latest research on ferroptosis, with a focus on its role in the pathogenesis of DBJDs and the therapeutic potential of natural products targeting this cell death pathway, offering novel insights for the prevention and treatment of DBJDs.
Collapse
Affiliation(s)
- Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Longlong Lv
- Weifang Hospital of Traditional Chinese Medicine, Weifang, China
| | - Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chenmoji Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiahao Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Daodi Qiu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haipeng Xue
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoqing Tan
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
41
|
Guo J, Li CG. Commentary: BAFF blockade attenuates DSS-induced chronic colitis via inhibiting NLRP3 inflammasome and NF-кB activation. Front Immunol 2024; 15:1528731. [PMID: 39776908 PMCID: PMC11703703 DOI: 10.3389/fimmu.2024.1528731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Affiliation(s)
- Jie Guo
- School of Medicine, Shenzhen University, Shenzhen, China
| | - Chen-guang Li
- School of Medicine, Shenzhen University, Shenzhen, China
- Pain Department of Shenzhen Nanshan People's Hospital, Shenzhen, China
| |
Collapse
|
42
|
Lim XR, Willemse L, Harraz OF. Amyloid beta Aβ 1-40 activates Piezo1 channels in brain capillary endothelial cells. Biophys J 2024:S0006-3495(24)04106-7. [PMID: 39722451 DOI: 10.1016/j.bpj.2024.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/12/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024] Open
Abstract
Amyloid beta (Aβ) peptide accumulation on blood vessels in the brain is a hallmark of neurodegeneration. While Aβ peptides constrict cerebral arteries and arterioles, their impact on capillaries is less understood. Aβ was recently shown to constrict brain capillaries through pericyte contraction, but whether-and if so how-Aβ affects endothelial cells (ECs) remains unknown. ECs represent the predominant vascular cell type in the cerebral circulation, and we recently showed that the mechanosensitive ion channel Piezo1 is functionally expressed in the plasma membrane of ECs. Since Aβ disrupts membrane structures, we hypothesized that Aβ1-40, the predominantly deposited isoform in the cerebral circulation, alters endothelial Piezo1 function. Using patch-clamp electrophysiology and freshly isolated capillary ECs, we assessed the impact of the Aβ1-40 peptide on single-channel Piezo1 activity. We show that Aβ1-40 increased Piezo1 open probability and channel open time. Aβ1-40 effects were absent when Piezo1 was genetically deleted or when a superoxide dismutase/catalase mimetic was used. Further, Aβ1-40 enhanced Piezo1 mechanosensitivity and lowered the pressure of half-maximal Piezo1 activation. Our data collectively suggest that Aβ1-40 facilitates higher Piezo1-mediated cation influx in brain ECs. These novel findings have the potential to unravel the possible involvement of Piezo1 modulation in the pathophysiology of neurodegenerative diseases characterized by Aβ accumulation.
Collapse
Affiliation(s)
- Xin Rui Lim
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont
| | - Luc Willemse
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont
| | - Osama F Harraz
- Department of Pharmacology, Larner College of Medicine, Vermont Center for Cardiovascular and Brain Health, University of Vermont, Burlington, Vermont.
| |
Collapse
|
43
|
Cordeiro BM, Leite Fontes CF, Meyer-Fernandes JR. Molecular Basis of Na, K-ATPase Regulation of Diseases: Hormone and FXYD2 Interactions. Int J Mol Sci 2024; 25:13398. [PMID: 39769162 PMCID: PMC11678576 DOI: 10.3390/ijms252413398] [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: 11/12/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The Na, K-ATPase generates an asymmetric ion gradient that supports multiple cellular functions, including the control of cellular volume, neuronal excitability, secondary ionic transport, and the movement of molecules like amino acids and glucose. The intracellular and extracellular levels of Na+ and K+ ions are the classical local regulators of the enzyme's activity. Additionally, the regulation of Na, K-ATPase is a complex process that occurs at multiple levels, encompassing its total cellular content, subcellular distribution, and intrinsic activity. In this context, the enzyme serves as a regulatory target for hormones, either through direct actions or via signaling cascades triggered by hormone receptors. Notably, FXYDs small transmembrane proteins regulators of Na, K-ATPase serve as intermediaries linking hormonal signaling to enzymatic regulation at various levels. Specifically, members of the FXYD family, particularly FXYD1 and FXYD2, are that undergo phosphorylation by kinases activated through hormone receptor signaling, which subsequently influences their modulation of Na, K-ATPase activity. This review describes the effects of FXYD2, cardiotonic steroid signaling, and hormones such as angiotensin II, dopamine, insulin, and catecholamines on the regulation of Na, K-ATPase. Furthermore, this review highlights the implications of Na, K-ATPase in diseases such as hypertension, renal hypomagnesemia, and cancer.
Collapse
Affiliation(s)
- Bárbara Martins Cordeiro
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - Carlos Frederico Leite Fontes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| | - José Roberto Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Rio de Janeiro 21941-590, RJ, Brazil
| |
Collapse
|
44
|
Hirata Y, Yamada Y, Taguchi S, Kojima R, Masumoto H, Kimura S, Niijima T, Toyama T, Kise R, Sato E, Uchida Y, Ito J, Nakagawa K, Taguchi T, Inoue A, Saito Y, Noguchi T, Matsuzawa A. Conjugated fatty acids drive ferroptosis through chaperone-mediated autophagic degradation of GPX4 by targeting mitochondria. Cell Death Dis 2024; 15:884. [PMID: 39643606 PMCID: PMC11624192 DOI: 10.1038/s41419-024-07237-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 12/09/2024]
Abstract
Conjugated fatty acids (CFAs) have been known for their anti-tumor activity. However, the mechanism of action remains unclear. Here, we identify CFAs as inducers of glutathione peroxidase 4 (GPX4) degradation through chaperone-mediated autophagy (CMA). CFAs, such as (10E,12Z)-octadecadienoic acid and α-eleostearic acid (ESA), induced GPX4 degradation, generation of mitochondrial reactive oxygen species (ROS) and lipid peroxides, and ultimately ferroptosis in cancer cell lines, including HT1080 and A549 cells, which were suppressed by either pharmacological blockade of CMA or genetic deletion of LAMP2A, a crucial molecule for CMA. Mitochondrial ROS were sufficient and necessary for CMA-dependent GPX4 degradation. Oral administration of an ESA-rich oil attenuated xenograft tumor growth of wild-type, but not that of LAMP2A-deficient HT1080 cells, accompanied by increased lipid peroxidation, GPX4 degradation and cell death. Our study establishes mitochondria as the key target of CFAs to trigger lipid peroxidation and GPX4 degradation, providing insight into ferroptosis-based cancer therapy.
Collapse
Affiliation(s)
- Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| | - Yuto Yamada
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Soma Taguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryota Kojima
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Haruka Masumoto
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Shinnosuke Kimura
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takuya Niijima
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takashi Toyama
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryoji Kise
- Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Emiko Sato
- Division of Clinical Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasunori Uchida
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Junya Ito
- Laboratory of Food Function Analysis, Graduate School of Agricultural Sciences, Tohoku University, Sendai, Japan
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Graduate School of Agricultural Sciences, Tohoku University, Sendai, Japan
| | - Tomohiko Taguchi
- Laboratory of Organelle Pathophysiology, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Asuka Inoue
- Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshiro Saito
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
45
|
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
|
46
|
Gowtham A, Chauhan C, Rahi V, Kaundal RK. An update on the role of ferroptosis in ischemic stroke: from molecular pathways to Neuroprotection. Expert Opin Ther Targets 2024; 28:1149-1175. [PMID: 39710973 DOI: 10.1080/14728222.2024.2446319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/29/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Ischemic stroke (IS), a major cause of mortality and disability worldwide, remains a significant healthcare challenge due to limited therapeutic options. Ferroptosis, a distinct iron-dependent form of regulated cell death characterized by lipid peroxidation and oxidative stress, has emerged as a crucial mechanism in IS pathophysiology. This review explores the role of ferroptosis in IS and its potential for driving innovative therapeutic strategies. AREA COVERED This review delves into the practical implications of ferroptosis in IS, focusing on molecular mechanisms like lipid peroxidation, iron accumulation, and their interplay with inflammation, reactive oxygen species (ROS), and the Nrf2-ARE antioxidant system. It highlights ferroptotic proteins, small-molecule inhibitors, and non-coding RNA modulators as emerging therapeutic targets to mitigate neuroinflammation and neuronal cell death. Studies from PubMed (1982-2024) were identified using MeSH terms such as 'Ferroptosis' and 'Ischemic Stroke,' and only rigorously screened articles were included. EXPERT OPINION Despite preclinical evidence supporting the neuroprotective effects of ferroptosis inhibitors, clinical translation faces hurdles such as suboptimal pharmacokinetics and safety concerns. Advances in drug delivery systems, bioinformatics, and AI-driven drug discovery may optimize ferroptosis-targeting strategies, develop biomarkers, and improve therapeutic outcomes for IS patients.
Collapse
Affiliation(s)
- A Gowtham
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Chandan Chauhan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Vikrant Rahi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Lucknow, India
| |
Collapse
|
47
|
Zhou J, Zhang L, Wu H, Gao SL, Chen XP, Zhang LF, Zhao CP, Wei BB, Bai Y. Ferroptosis-related lncRNA AL136084.3 is associated with NUPR1 in bladder cancer. Discov Oncol 2024; 15:730. [PMID: 39613992 DOI: 10.1007/s12672-024-01564-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/11/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND LncRNAs are critical regulators of bladder cancer (BLCA), and ferroptosis is a newly discovered cell death responsible for mediating apoptosis and tumorigenesis. The present study aims to establish a prognostic signature of differentially expressed ferroptosis-related lncRNAs (DEFRlncRNAs) and explore the DEFRlncRNA associated with NUPR1 in BLCA. METHODS DEFRlncRNAs in BLCA patients were screened using univariate and multivariate Cox and LASSO regression analyses. In vitro experiments were performed to detect the regulatory effects of DEFRlncRNAs on BLCA cells. A prognostic signature of DEFRlncRNAs in BLCA was created and validated. Moreover, we used RNA-binding protein immunoprecipitation (RIP) to evaluate the correlated DEFRlncRNA with NUPR1. RESULTS A prognostic signature involving 18 DEFRlncRNAs in BLCA was created. Overexpression of AL355353.2 or knockdown of AL136084.3 promoted apoptosis in 5637 and T24 cells in vitro. Results from Starbase database estimated that AL136084.3 was positively associated with NUPR1 (R = 0.229, p < 0.001). RIP analysis revealed the reciprocal binding of NUPR1 and AL136084.3 in BLCA. CONCLUSION The identified FRlncRNA pair signature has a good prognostic and clinical predictive value. The ferroptosis-related lncRNA AL136084.3 is correlated with NUPR1 in BLCA.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Hefeng Road 1000, Wuxi, 214000, China
| | - Li Zhang
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China
| | - Hao Wu
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China
- Department of Urology, Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, Zhejiang, China
| | - Sheng-Lin Gao
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China
| | - Xiao-Ping Chen
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Hefeng Road 1000, Wuxi, 214000, China
| | - Li-Feng Zhang
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China
| | - Cui-Ping Zhao
- Department of Geriatrics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, China.
| | - Bing-Bing Wei
- Department of Urology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, 299 Qingyang Road, Wuxi, 214023, China.
| | - Yu Bai
- Department of Urology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 68 Gehu Road, Changzhou, 213000, Jiangsu, China.
- Department of Urology, Gonghe County Traditional Chinese Medicine Hospital, 277 South Street of Qinghai Lake, Gonghe County, Hainan Prefecture, 813099, Qinghai, China.
| |
Collapse
|
48
|
Qi WH, Tang N, Zhao ZJ, Li XQ. Transient receptor potential channels in viral infectious diseases: Biological characteristics and regulatory mechanisms. J Adv Res 2024:S2090-1232(24)00541-1. [PMID: 39551130 DOI: 10.1016/j.jare.2024.11.022] [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: 06/15/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Viral infectious diseases have long posed a challenge to humanity. In recent decades, transient receptor potential (TRP) channels have emerged as newly investigated cation channels. Increasing evidence suggests that TRP channel-mediated Ca2+ homeostasis disruptions, along with associated pathological changes, are critical factors in the onset and progression of viral infectious diseases. However, the precise roles and mechanisms of TRP channels in these diseases remain to be systematically elucidated. AIM OF REVIEW The aim of this review is to systematically summarize recent advances in understanding TRP channels in viral infections, and based on current progress and challenges, propose future directions for research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review summarizes the classification and biological functions of the TRP family, explores the mechanisms by which TRP channels contribute to viral infections, and highlights specific mechanisms at three levels: virus, host, and outcome. These include the direct role in viral biology and replication, the indirect role in host immunity and inflammation, and the resulting pathological changes. Additionally, we discuss the potential applications of the TRP family in the treatment of viral infectious diseases and propose future research directions.
Collapse
Affiliation(s)
- Wen-Hui Qi
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, Air Force Medical University, Xi'an, Shaanxi 710032, China; Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Na Tang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, Air Force Medical University, Xi'an, Shaanxi 710032, China; Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Zhi-Jing Zhao
- Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China; Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Xiao-Qiang Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, Air Force Medical University, Xi'an, Shaanxi 710032, China; Research Institution, Xijing Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
49
|
Park VS, Pope LE, Ingram J, Alchemy GA, Purkal J, Andino-Frydman EY, Jin S, Singh S, Chen A, Narayanan P, Kongpachith S, Phillips DC, Dixon SJ, Popovic R. Lipid composition differentiates ferroptosis sensitivity between in vitro and in vivo systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.622381. [PMID: 39605501 PMCID: PMC11601366 DOI: 10.1101/2024.11.14.622381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Ferroptosis is a regulated non-apoptotic cell death process characterized by iron-dependent lipid peroxidation. This process has recently emerged as a promising approach for cancer therapy. Peroxidation of polyunsaturated fatty acid-containing phospholipids (PUFA-PLs) is necessary for the execution of ferroptosis. Ferroptosis is normally suppressed by glutathione peroxidase 4 (GPX4), which reduces lipid hydroperoxides to lipid alcohols. Some evidence indicates that GPX4 may be a useful target for drug development, yet factors that govern GPX4 inhibitor sensitivity in vivo are poorly understood. We find that pharmacological and genetic loss of GPX4 function was sufficient to induce ferroptosis in multiple adherent ("2D") cancer cell cultures. However, reducing GPX4 protein levels did not affect tumor xenograft growth when these cells were implanted in mice. Furthermore, sensitivity to GPX4 inhibition was markedly reduced when cells were cultured as spheroids ("3D"). Mechanistically, growth in 3D versus 2D conditions reduced the abundance of PUFA-PLs. 3D culture conditions upregulated the monounsaturated fatty acid (MUFA) biosynthetic gene stearoyl-CoA desaturase (SCD). SCD-derived MUFAs appear to protect against ferroptosis in 3D conditions by displacing PUFAs from phospholipids. Various structurally related long chain MUFAs can inhibit ferroptosis through this PUFA-displacement mechanism. These findings suggest that growth-condition-dependent lipidome remodeling is an important mechanism governing GPX4 inhibitor effects. This resistance mechanism may specifically limit GPX4 inhibitor effectiveness in vivo .
Collapse
|
50
|
De Leon-Oliva D, Boaru DL, Minaya-Bravo AM, De Castro-Martinez P, Fraile-Martinez O, Garcia-Montero C, Cobo-Prieto D, Barrena-Blázquez S, Lopez-Gonzalez L, Albillos A, Alvarez-Mon M, Saez MA, Diaz-Pedrero R, Ortega MA. Improving understanding of ferroptosis: Molecular mechanisms, connection with cellular senescence and implications for aging. Heliyon 2024; 10:e39684. [PMID: 39553553 PMCID: PMC11564042 DOI: 10.1016/j.heliyon.2024.e39684] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
In the face of cell damage, cells can initiate a response ranging from survival to death, the balance being crucial for tissue homeostasis and overall health. Cell death, in both accidental and regulated forms, plays a fundamental role in maintaining tissue homeostasis. Among the regulated mechanisms of cell death, ferroptosis has garnered attention for its iron-dependent phospholipid (PL) peroxidation and its implications in aging and age-related disorders, as well as for its therapeutic relevance. In this review, we provide an overview of the mechanisms, regulation, and physiological and pathological roles of ferroptosis. We present new insights into the relationship between ferroptosis, cellular senescence and aging, emphasizing how alterations in ferroptosis pathways contribute to aging-related tissue dysfunction. In addition, we examine the therapeutic potential of ferroptosis in aging-related diseases, offering innovative insights into future interventions aimed at mitigating the effects of aging and promoting longevity.
Collapse
Affiliation(s)
- Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Ana M. Minaya-Bravo
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - David Cobo-Prieto
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Immune System Diseases-Rheumatology Service, Central University Hospital of Defence-UAH Madrid, 28801, Alcala de Henares, Spain
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala de Henares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Agustín Albillos
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Gastroenterology and Hepatology Service, Ramón y Cajal University Hospital, University of Alcalá, IRYCIS, Network Biomedical Research Center for Liver and Digestive Diseases (CIBERehd), Carlos III Health Institute, Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806, Alcala de Henares, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, 28801, Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, Alcala de Henares, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, (CIBERehd), Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| |
Collapse
|