1
|
Zhang DY, Li D, Chen SJ, Zhang LJ, Zhu XL, Chen FD, Chen C, Wang Q, Du Y, Xiong JX, Huang SM, Zhang XD, Lv YT, Zeng F, Chen RX, Huang X, Mao F, Zhou S, Yao Q, Huang Y, Chen R, Mo Y, Xie Y, Jiang YH, Chen Z, Mo CY, Chen JJ, Bai FH. Bacteroides uniformis-generated hexadecanedioic acid ameliorates metabolic-associated fatty liver disease. Gut Microbes 2025; 17:2508433. [PMID: 40413726 PMCID: PMC12118425 DOI: 10.1080/19490976.2025.2508433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/28/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025] Open
Abstract
Gut microbiota exerts a pivotal influence on the development of Metabolic Associated Fatty Liver Disease (MAFLD), although the specific contributions of individual bacterial strains and their metabolites remain poorly defined. We conducted stool shotgun metagenomic sequencing and plasma untargeted metabolomics in a large prospective cohort comprising 120 MAFLD patients and 120 matched healthy controls. The mechanisms and microbial-derived metabolites involved in MAFLD were further investigated through multi-omics analyses in vitro and in vivo. Distinct differences were identified in both the microbial community structure and metabolomic profiles between MAFLD patients and healthy controls. Bacteroides uniformis (B. uniformis) was the most significantly depleted species in MAFLD and negatively correlated with hepatic steatosis and BMI. MAFLD was characterized by marked disruptions in fatty acid and amino acid metabolism. Combined analysis of metabolomic and metagenomic data achieved high diagnostic accuracy for MAFLD and hepatic steatosis severity (AUC = 0.93). Transplantation of fecal microbiota from MAFLD subjects into ABX mice led to the onset of MAFLD-like symptoms, whereas B. uniformis administration alleviate disease progression by inhibiting intestinal fat absorption, FFA from eWAT influx into liver via the gut-liver axis, and IRE1α-XBP1s-mediated flipogenesis and ferroptosis, as confirmed by hepatic transcriptomic and proteomic analyses. Hexadecanedioic acid (HDA), potentially identified as a key metabolite produced by B. uniformis, ameliorated MAFLD symptoms. Mechanistically, B. uniformis-derived HDA also inhibited fat absorption and transported, and entered the liver via the portal vein to suppress IRE1α-XBP1s-mediated flipogenesis and ferroptosis. B. uniformis and its potential putative metabolite HDA may contribute to MAFLD progression modulation, through regulation of the IRE1α-XBP1s axis. This study provides new insights into the gut-liver axis in MAFLD and offers promising therapeutic targets based on specific microbes and their metabolites.
Collapse
Affiliation(s)
- Da-Ya Zhang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Da Li
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shi-Ju Chen
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Li-Jun Zhang
- Health Management Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xu-Li Zhu
- Department of Gastroenterology, Otog Front Banner People’s Hospital, Otog Front Banner, China
| | - Fa-Di Chen
- Wuzhishan Center for Disease Control and Prevention, Wuzhishan, China
| | - Chen Chen
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qi Wang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yiping Du
- Cardiovascular Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jian-Xin Xiong
- Department of Gastroenterology, Hainan Second People’s Hospital, Wuzhishan, China
| | - Shi-Mei Huang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xiao-Dong Zhang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yan-Ting Lv
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Fan Zeng
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Run-Xiang Chen
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xianfeng Huang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Fengjiao Mao
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shuo Zhou
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qicen Yao
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yuliang Huang
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Runyu Chen
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ying Mo
- The Second School of Clinical Medicine, Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yunqian Xie
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yue-Hong Jiang
- Department of Gastroenterology, The Second People’s Hospital of Ledong Li Autonomous County, Ledong Li Autonomous County, China
| | - Zhai Chen
- Department of Gastroenterology, Dongfang People’s Hospital, Dongfang, China
| | - Cui-Yi Mo
- Department of Gastroenterology, Qionghai People’s Hospital, Qionghai, China
| | - Jia-Jia Chen
- Department of Gastroenterology, Qionghai People’s Hospital, Qionghai, China
| | - Fei-Hu Bai
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
- Department of Gastroenterology, The Gastroenterology Clinical Medical Center of Hainan Province, Haikou, China
| |
Collapse
|
2
|
Shao C, Xia W, Liu Y. Bioinformatic Analysis and Molecular Docking Identify Isorhamnetin Is a Candidate Compound in the Treatment of Pulmonary Artery Hypertension. Anatol J Cardiol 2025; 29:52-65. [PMID: 39605239 PMCID: PMC11793806 DOI: 10.14744/anatoljcardiol.2024.4723] [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: 08/03/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The current study aims to identify the key pathways and potential therapeutic targets for pulmonary arterial hypertension (PAH) and to further evaluate the anti-PAH effects of isorhamnetin. METHODS The dataset of gene expression profiling for PAH (GSE113439) was downloaded from the gene expression omnibus (GEO) database. Isorhamnetin target genes were extracted from the comparative toxicogenomics database (CTD). Various bioinformatics methods were employed to identify the core pathways associated with PAH and potential intervention targets. Molecular docking was conducted between the interacting target and the candidate compound, isorhamnetin. RESULTS One thousand nine hundred sixty-two upregulated genes and 642 downregulated genes were identified. Molecular complex detection analyses revealed that the significant biological processes associated with upregulated genes included DNA damage response, mitotic cell cycle, and chromosome organization. In contrast, the signifi ant biological processes related to downregulated genes encompassed cellular response to growth factor stimulus, response to growth factor, and blood vessel development. Immune infilt ation analysis indicated that PAH is associated with signifi ant changes in the distribution of immune cells and differential expression of immune checkpoints. Furthermore, 58 isorhamnetin targets were extracted from the CTD, and we identified 1 interacting gene, NFE2L2, among the differentially expressed genes (DEGs), DEGs related to ferroptosis, and isorhamnetin targets. Isorhamnetin demonstrated strong affinities with vascular endothelial growth factor (VEGF) receptors and transcription factors (ATM and ZNF24) associated with VEGFs, as well as the ferroptosis protein NFE2L2. CONCLUSIONS Pulmonary arterial hypertension is characterized by a series of abnormalities in downstream molecular signaling pathways, including DNA damage, immune dysregulation, VEGF signaling deficienc , and the ferroptosis process. These may represent the core pathophysiological mechanisms of PAH. Ferroptosis-related genes, such as NFE2L2 and TF (ATM, ZNF24) associated with VEGFs, are potential therapeutic targets that contribute to the mechanisms mentioned above. Isorhamnetin is a promising candidate compound for the treatment of PAH.
Collapse
Affiliation(s)
- Chen Shao
- Department of Nursing Science, The Second People’s Hospital of Lianyungang, Jiangsu, China
| | - Wei Xia
- Department of Pharmacology, The Second People’s Hospital of Lianyungang, Jiangsu, China
| | - Yang Liu
- Department of Internal and Pediatrics, School of Clinical Medicine, Qilu Medical University, Zibo, Shandong, 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
|
Yang L, Xiao JJ, Zhang L, Lu Q, Hu BB, Liu Y, Pu JX, Hu JW, Yu H, Wu X, Zhang BF. Methionine sulfoxide reductase A deficiency aggravated ferroptosis in LPS-induced acute kidney injury by inhibiting the AMPK/NRF2 axis and activating the CaMKII/HIF-1α pathway. Free Radic Biol Med 2025; 234:248-263. [PMID: 40288699 DOI: 10.1016/j.freeradbiomed.2025.04.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/13/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Methionine sulfoxide reductase A (MsrA) is an important antioxidant enzyme that is present in various tissues and play a crucial role in many pathological processes. However, the role of MsrA in acute kidney injury (AKI) requires further exploration. Here, we aimed to explore whether MsrA is involved in sepsis-associated AKI and the underlying mechanisms. In the present study, AKI was induced by lipopolysaccharide (LPS) in WT mice and MsrA knockout mice. The role of MsrA in LPS-induced injury in the human renal proximal tubule epithelial cell line HK-2 was also examined by MsrA knockdown. MsrA deficiency exacerbated LPS-induced kidney damage in vivo. In addition, MsrA deficiency and silencing intensified iron overload, lipid peroxidation and ferroptosis in LPS-stimulated renal tubular cells. The mechanistic study revealed that MsrA knockout or knockdown led to the oxidation of calcium/calmodulin-dependent protein kinase II (CaMKII) at methionine 281/282, resulting in sustained activation of CaMKII, which upregulated iron metabolism-related proteins such as transferrin receptor 1 (TFR1) by promoting phosphorylation and nuclear translocation of hypoxia-inducible factor-1α (HIF-1α) and induced abnormal iron metabolism. Meanwhile, CaMKII activation downregulated the expression of glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11) by inhibiting the activity of AMP-activated protein kinase (AMPK) and phosphorylation of nuclear factor erythroid 2-related factor 2 (NRF2), resulting in lipid peroxidation. Consequently, LPS-induced ferroptosis was exacerbated. Our study is the first to reveal that MsrA deficiency intensifies LPS-induced ferroptosis through CaMKII activation in renal tubular cells. There are two major mechanisms: one is the promotion of lipid peroxidation by inhibiting the AMPK/NRF2 axis, and the other is abnormal iron metabolism by activating the HIF-1α/TFR1 pathway. MsrA may be a potential therapeutic target for organ and cell damage induced by ferroptosis.
Collapse
Affiliation(s)
- LiJiao Yang
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jing-Jie Xiao
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China; Department of Cardiology, Zhongnan Hospital of Wuhan University, Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, 430071, China
| | - Lian Zhang
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - QianYu Lu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bin-Bin Hu
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Yu Liu
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Jun-Xing Pu
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Jun-Wei Hu
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Hong Yu
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China.
| | - XiaoYan Wu
- Department of Nephrology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Bai-Fang Zhang
- Department of Biochemistry and Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China.
| |
Collapse
|
5
|
Gao D, Chen T, Dong J, He Y, Ge N, Guo J, Sun S, Yang F. Ferroptosis at the crossroads: Insights and advances in non-neoplastic pancreatic diseases. Int Immunopharmacol 2025; 158:114870. [PMID: 40383100 DOI: 10.1016/j.intimp.2025.114870] [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: 12/11/2024] [Revised: 03/26/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
Ferroptosis is a form of regulated cell death characterized by iron accumulation and increased lipid peroxidation, primarily counteracted by a range of antioxidant molecules, including glutathione (GSH), glutathione peroxidase 4 (GPX4), ubiquinone, tetrahydrofolate, and nuclear respiratory factor 2. Furthermore, the process of ferroptosis is intricately influenced by the opposing actions of the p53 tumor suppressor gene and activated transcription factors 3 and 4, which can either facilitate or hinder ferroptotic cell death depending on the cellular context. This form of cell death is significantly associated with various pancreatic disorders, including both acute and chronic pancreatitis, as well as diabetes mellitus. In this review, we thoroughly investigate the mechanisms underlying ferroptosis, focusing on iron overload, lipid peroxidation, and the regulatory molecules involved in ferroptosis modulation (notably the system xc-/GSH/GPX4 axis), along with the relevant signaling pathways. We also examine the role of ferroptosis in non-neoplastic pancreatic diseases such as pancreatitis and diabetes mellitus while identifying novel therapeutic agents that target ferroptosis, potentially paving the way for innovative treatment strategies for pancreatic conditions.
Collapse
Affiliation(s)
- Duolun Gao
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Tingting Chen
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Jize Dong
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Yanjie He
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York 10012, NY, USA
| | - Nan Ge
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Jintao Guo
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Siyu Sun
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China.
| | - Fan Yang
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China.
| |
Collapse
|
6
|
Gao J, Li X, Hou Y, Li Y, Pang Y, Wu X, Zhao L, Zhang J, Wang H, Wang H, Dong J, Xu X, Peng R, Wang Y, Yao B. Microwave exposure induced ferroptosis by inhibiting the Nrf2 pathway and affected reproductive function in male mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 301:118523. [PMID: 40527021 DOI: 10.1016/j.ecoenv.2025.118523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 06/11/2025] [Accepted: 06/12/2025] [Indexed: 06/19/2025]
Abstract
The mechanisms underlying the negative health effects of microwave exposure on male reproduction remain unclear. Thus, this study aimed to explore the role and regulatory mechanisms of ferroptosis in microwave-induced reproductive damage. The exposure of male C57 mice to 2.856 GHz of microwave radiation at 0, 10 and 20 mW/cm² for 30 min decreased sperm motility, induced morphological changes, damaged testicular tissue and mitochondrial morphology, increased malondialdehyde (MDA) contents and decreased the GSH/GSSG ratio. Simultaneously, the Fe²⁺ levels increased and SLC7A11 and GPX4 protein expressions decreased, causing oxidative stress. After 30 min of mouse spermatocyte (GC-2) irradiation, the cell viability of the Fer-1 inhibitor group and the GSH/GSSG ratio increased, while the reactive oxygen species, MDA and ferrous iron contents decreased. Furthermore, the depolarisation of membrane potential improved. Western blotting revealed that Nrf2, Keap1, SLC7A11, GPX4 and HO-1 expressions were down-regulated by microwave exposure and significantly up-regulated following the addition of the Fer-1 inhibitor. The results confirmed that the Nrf2 signalling pathway can regulate ferroptosis of oxidative stress. This study demonstrates that microwave exposure affects mouse reproductive function by enhancing oxidative stress, inducing ferroptosis by inhibiting the Nrf2 signalling pathway and reducing SLC7A11 and GPX4 protein expressions.
Collapse
Affiliation(s)
- Jingchao Gao
- Beijing Institute of Radiation Medicine, Beijing 100850, China; Department of Oncology, Air Force Medical Center of the PLA, Beijing 100142, China.
| | - Xinyue Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Public Health, Wannan Medical College, Wuhu 241002, China.
| | - Yizhuo Hou
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Yanyang Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Yueyue Pang
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Xiaoran Wu
- Beijing Institute of Radiation Medicine, Beijing 100850, China; School of Public Health, Wannan Medical College, Wuhu 241002, China.
| | - Li Zhao
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Jing Zhang
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Haoyu Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Hui Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Ji Dong
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Xinping Xu
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Ruiyun Peng
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Yu Wang
- Department of Oncology, Air Force Medical Center of the PLA, Beijing 100142, China.
| | - Binwei Yao
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
7
|
Huang JY, Yu HN. The role of the Nrf2 pathway in inhibiting ferroptosis in kidney disease and its future prospects. Pathol Res Pract 2025; 272:156084. [PMID: 40527053 DOI: 10.1016/j.prp.2025.156084] [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: 03/20/2025] [Revised: 06/12/2025] [Accepted: 06/12/2025] [Indexed: 06/19/2025]
Abstract
Kidney disease (KD) has gradually become a major social and economic burden on the healthcare system. Recent studies highlight ferroptosis as a critical mechanism in the progression of these conditions. Recognized for its essential role in renal pathogenesis, ferroptosis is attracting increasing research attention and emerging as a key focus of investigation. The Nrf2 signaling pathway, known for its regulatory influence on ferroptosis, plays a central role in this context. Activation of the nuclear factor E2-related factor 2 (Nrf2) pathway and the subsequent attenuation of ferroptosis present substantial opportunities as novel therapeutic targets for managing kidney injury (KI). This article summarizes the latest mechanism of action of the Nrf2 pathway in ferroptosis, explores how the Nrf2 pathway affects ferroptosis in KD therapy, and investigates the potential therapeutic effects of natural products targeting the Nrf2 pathway. These natural products have been shown to inhibit ferroptosis through the Nrf2 pathway, providing new insights and therapeutic strategies for the clinical and individualized management of KD.
Collapse
Affiliation(s)
- Jia-Yuan Huang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Hai-Ning Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China.
| |
Collapse
|
8
|
Bai X, Duan T, Shao J, Zhang Y, Xing G, Wang J, Liu X, Wang M, He Y, Wang H, Zhang ZY, Ni M, Zhou JY, Pan J. CBX3 promotes multidrug resistance by suppressing ferroptosis in colorectal carcinoma via the CUL3/NRF2/GPX2 axis. Oncogene 2025; 44:1678-1693. [PMID: 40089640 PMCID: PMC12122363 DOI: 10.1038/s41388-025-03337-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 02/01/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Chemoresistance poses a significant challenge in colorectal cancer (CRC) treatment. However, the mechanisms underlying chemoresistance remain unclear. CBX3 promoted proliferation and metastasis in CRC. However, the role and mechanism of CBX3 in chemoresistance remain unknown. Therefore, we aimed to investigate the effects and mechanisms of CBX3 on multidrug resistance in CRC. Our studies showed that higher levels of CBX3 expression were associated with poor survival, especially in groups with progression following chemotherapy. CBX3 overexpression increased Irinotecan and Oxaliplatin resistance, whereas CBX3 knockdown suppressed multidrug resistance in CRC cells. Additionally, CBX3 inhibited ferroptosis associated with multidrug resistance, and the ferroptosis activators prevented CBX3 overexpression-mediated cell survival. RNA sequencing revealed that the NRF2-signaling pathway was involved in this process. CBX3-upregulated NRF2 protein expression by directly binding to the promoter of Cullin3 (CUL3) to suppress CUL3 transcription and CUL3-mediated NRF2 degradation. Moreover, Glutathione Peroxidase 2 (GPX2) was downstream of the CBX3-NRF2 pathway in CRC chemoresistance. ML385, an NRF2 inhibitor, suppressed GPX2 expression, and increased ferroptosis in PDX models. Our study identified CBX3/NRF2/GPX2 axis may be a novel signaling pathway that mediates multidrug resistance in CRC. This study proposes developing novel strategies for cancer treatment to overcome drug resistance in the future.
Collapse
Affiliation(s)
- Xiaoming Bai
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Tinghong Duan
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
- Department of Pathology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, PR China
| | - Jiaofang Shao
- Department of Bioinformatics, Nanjing Medical University, Nanjing, PR China
| | - Yutong Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Guangyuan Xing
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Jie Wang
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Xue Liu
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Min Wang
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Yuanqiao He
- Center of Laboratory Animal Science, Nanchang University, Nanchang, PR China
- Key Laboratory of New Drug Evaluation and Transformation of Jiangxi Province, Nanchang Royo Biotech Co., Ltd, Nanchang, PR China
| | - Hai Wang
- Department of Pathology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, PR China
| | - Zhi-Yuan Zhang
- Department of Pathology, Nanjing Medical University, Nanjing, PR China
| | - Min Ni
- Department of Colorectum, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, PR China.
| | - Jin-Yong Zhou
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, PR China.
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, PR China.
| | - Jinshun Pan
- Department of Biotherapy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
9
|
Sekine H, Akaike T, Motohashi H. Oxygen needs sulfur, sulfur needs oxygen: a relationship of interdependence. EMBO J 2025; 44:3307-3326. [PMID: 40394395 DOI: 10.1038/s44318-025-00464-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/22/2025] Open
Abstract
Oxygen and sulfur, both members of the chalcogen group (group 16 elements), play fundamental roles in life. Ancient organisms primarily utilized sulfur for energy metabolism, while the rise in atmospheric oxygen facilitated the evolution of aerobic organisms, enabling highly efficient energy production. Nevertheless, all modern organisms, both aerobes and anaerobes, must protect themselves from oxygen toxicity. Interestingly, aerobes still rely on sulfur for survival. This dependence has been illuminated by the recent discovery of supersulfides, a novel class of biomolecules, made possible through advancements in technology and analytical methods. These breakthroughs are reshaping our understanding of biological processes and emphasizing the intricate interplay between oxygen and sulfur in regulating essential redox reactions. This review summarizes the latest insights into the biological roles of sulfur and oxygen, their interdependence in key processes, and their contributions to adaptive responses to environmental stressors. By exploring these interactions, we aim to provide a comprehensive perspective on how these elements drive survival strategies across diverse life forms, highlighting their indispensable roles in both human health and the sustenance of life.
Collapse
Affiliation(s)
- Hiroki Sekine
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Takaaki Akaike
- Department of Redox Molecular Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
10
|
Zhao Y, Liang H, Cui X. Obacunone regulates ferroptosis in ovarian cancer through the Akt/p53 pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:7027-7039. [PMID: 39708098 DOI: 10.1007/s00210-024-03738-9] [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: 10/16/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Ovarian cancer is characterized by a high rate of recurrence and a poor prognosis. Ferroptosis, a programmed cell death that is dependent on iron and lipid peroxidation, has emerged as a novel therapeutic target in recent years. This study investigated the effects of Obacunone, a naturally occurring compound present in citrus fruits, on the induction of ferroptosis in ovarian cancer via the Akt/p53 signaling pathway. SKOV3 and OVCAR3 ovarian cancer cell lines were utilized in vitro, while a BALB/c nude mouse model was employed for in vivo experiments. Cell proliferation was assessed utilizing the CCK-8 assay and EDU incorporation. The western blot technique was employed to assess the expression levels of proteins associated with the Akt/p53 signaling pathway. The ferroptosis inhibitor Fer-1 and the Akt activator SC79 were utilized to investigate the potential mechanism of action of Obacunone. Obacunone significantly inhibited the proliferation of ovarian cancer cells and induced ferroptosis, as evidenced by increased intracellular iron content, elevated lipid peroxidation levels, and abnormal mitochondrial morphology. Obacunone also decreased GSH levels, inhibited GPX4 expression and up-regulated ACSL4, as well as reduced Akt phosphorylation and enhanced p53 expression. In vivo experiments showed that Obacunone effectively inhibited tumor growth. Obacunone exhibits potential therapeutic significance through the modulation of the Akt/p53 signaling pathway, which may induce ferroptosis and inhibit the proliferation of ovarian cancer cells.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Traditional Chinese Medicine, Yantaishan Hospital, Yantai, 264000, Shandong, China
| | - Haiyue Liang
- Department of Drug Business Management, Yantai Center for Food and Drug Control, Yantai, 264000, Shandong, China
| | - Xinmu Cui
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, No.39 Xingfu Road, Zhifu District, Yantai City, 264000, Shandong Province, China.
| |
Collapse
|
11
|
Alarcón-Veleiro C, López-Calvo I, Berjawi L, Lucio-Gallego S, Mato-Basalo R, Quindos-Varela M, Lesta-Mellid R, Santamarina-Caínzos I, Varela-Rodríguez S, Fraga M, Quintela M, Vizoso-Vázquez A, Arufe MC, Fafián-Labora J. Ferroptosis: An emerging strategy for managing epithelial ovarian cancer. Biomed Pharmacother 2025; 187:118065. [PMID: 40306179 DOI: 10.1016/j.biopha.2025.118065] [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: 01/09/2025] [Revised: 03/30/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
Ferroptosis is a regulated form of cell death characterised by iron-dependent lipid peroxidation, a process intricately linked to cellular redox homeostasis. This form of cell death is induced by the accumulation of intracellular iron and the subsequent generation of reactive oxygen species (ROS), which leads to lipid peroxidation and ultimately cell death. Ferroptosis is distinct from traditional forms of cell death, such as apoptosis, and holds significant therapeutic potential, particularly in cancers harboring rat sarcoma virus (RAS) mutations, such as epithelial ovarian cancer (EOC). EOC is notoriously resistant to conventional therapies and is associated with a poor prognosis. In this review, we examine recent progress in the understanding of ferroptosis, with a particular focus on its redox biology and the complex regulatory networks involved. We also propose a novel classification system for ferroptosis modulators, grouping them into six categories (I, II, III, IV, V and VI) based on their mechanisms of action and their roles in modulating cellular redox status. By refining these categories, we aim to provide deeper insights into the role of ferroptosis in cancer biology, especially in EOC, and to identify potential therapeutic avenues. We propose that further investigation of ferroptosis in the context of redox biology could reveal novel biomarkers and therapeutic targets, offering promising strategies to overcome resistance mechanisms and improve clinical outcomes for patients with EOC and other treatment-resistant cancers.
Collapse
Affiliation(s)
- C Alarcón-Veleiro
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - I López-Calvo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain; Centro Interdisciplinar de Química de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Rúa as Xubias 84, A Coruña 15006, Spain
| | - L Berjawi
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - S Lucio-Gallego
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - R Mato-Basalo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - M Quindos-Varela
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - R Lesta-Mellid
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - I Santamarina-Caínzos
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - S Varela-Rodríguez
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - M Fraga
- Department of Anatomical Pathology, University Hospital Complex A Coruña, As Xubias 84, A Coruña 15006, Spain
| | - M Quintela
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - A Vizoso-Vázquez
- Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain
| | - M C Arufe
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| | - J Fafián-Labora
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| |
Collapse
|
12
|
Han Z, Han S, Fang X, Lu M, Mao Y, Shi L, Song J, Wang T, Xiao J, Xiang L, Yang C, Zhu Z, Wang Y, Feng J. Acetyl-CoA carboxylase activation disrupts iron homeostasis to drive ferroptosis. Free Radic Biol Med 2025:S0891-5849(25)00720-8. [PMID: 40449808 DOI: 10.1016/j.freeradbiomed.2025.05.421] [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: 03/05/2025] [Revised: 05/20/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
Acetyl-CoA carboxylase (ACC) is a rate-limiting enzyme in de novo lipogenesis. Here, we show a unique function of ACC in disrupting cellular iron homeostasis to drive ferroptosis, an iron-dependent, lipid peroxidation-induced form of cell death. We observed neuronal lipid accumulation and elevated labile iron pool associated with ACC dephosphorylation in mouse models of obstructive sleep apnea (OSA), a highly prevalent neurodegenerative disorder. ACC gene (Acaca) knockout (KO) or inhibition of its enzymatic activity rescued cellular iron metabolism through restoring lysosomal integrity and function, suppressing neuronal ferroptosis. ACC inactivation-driven lysosomal iron homeostasis requires the NFE2L2/NRF2-TFEB axis. Empagliflozin mitigates cellular iron overload via the ACC-NRF2-TFEB-lysosome pathway to alleviate neuronal ferroptosis, cognitive impairment, and mood dysfunction in OSA mice. Furthermore, inhibiting neuronal ACC reduces microglial activation, characterized by elevated complement proteins and pro-inflammatory cytokines, while microglia-specific C1qa KO prevents neuronal injury in OSA mice. Our findings identify a unique coupling between iron homeostasis and lipogenic signaling, suggesting ACC as a potential therapeutic target for neuronal ferroptosis and the resultant microgliosis in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ziqi Han
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Shuangyu Han
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Xiaoyan Fang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Mengyu Lu
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Yuanling Mao
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Leilei Shi
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Junxiu Song
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Tian Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Jichen Xiao
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Li Xiang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Changqing Yang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Zhigang Zhu
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital
| | - Yubao Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital.
| | - Jing Feng
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital.
| |
Collapse
|
13
|
Hu C, Gao S, Li X, Yang K, Cheng Y, Guo W, Wu H, Cheng X, Zhao W, Kong Y, Hu H, Wang S. Crosstalk of autophagy and ferroptosis in cardiovascular diseases: from pathophysiology to novel therapy. Redox Biol 2025; 84:103705. [PMID: 40450834 PMCID: PMC12164230 DOI: 10.1016/j.redox.2025.103705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/19/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Cardiovascular diseases (CVDs) are characterized by high morbidity and mortality rates, imposing substantial epidemiological and economic burdens worldwide. Among the multifaceted mechanisms implicated in CVDs, autophagy and ferroptosis, two intimately linked cellular processes, emerge as pivotal pathophysiological contributors. Autophagy, as an evolutionary conserved process that mediates the degradation and recycling of intracellular components, including proteins and organelles, exerts critical regulatory effects on iron metabolism and lipid homeostasis through various specialized forms, including ferritinophagy and lipophagy. Conversely, ferroptosis, an iron dependent form of cell death, involves oxidative stress and the accumulation of lipid peroxides, often triggered by iron overload and the dysfunction of glutathione peroxidase 4 (GPX4). The intricate crosstalk between these two processes, particularly ferritinophagy-mediated iron regulation influencing ferroptosis, plays a crucial role in diverse CVDs contexts. Key regulatory molecules, such as Beclin-1 and nuclear factor E2-related factor 2 (Nrf2), function as central hubs, orchestrating the intricate interplay between autophagy and ferroptosis. Through a comprehensive examination of these mechanisms across various CVDs pathologies, we summarize the latest findings and outline potential therapeutic strategies targeting the crosstalk between autophagy and ferroptosis. As the inaugural review focusing on autophagy-ferroptosis interactions in CVDs, this work significantly enriches our understanding of the pathophysiology of CVDs and identifies novel therapeutic targets with potential for precision medicine interventions in managing CVDs.
Collapse
Affiliation(s)
- Changhao Hu
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Siying Gao
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Xinyi Li
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Kaiqing Yang
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Ye Cheng
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Wei Guo
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Huijun Wu
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Xueqin Cheng
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Weiwen Zhao
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Yuxuan Kong
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China
| | - Haoyuan Hu
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China.
| | - Songyun Wang
- Cardiovascular Hospital, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Center of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, China.
| |
Collapse
|
14
|
Yuan J, Huang W, Lin M, Sun S, Zhong F, Ye L, Yin H, Ou X, Zeng Z. ML385 increases ferroptosis via inhibiting Nrf2/HO-1 pathway to enhances the sensitivity of MCF-7 TAMR to tamoxifen. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04256-y. [PMID: 40425765 DOI: 10.1007/s00210-025-04256-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025]
Abstract
The purpose of this study was to study the mechanism of the increased sensitivity of breast cancer tamoxifen-resistant strains by enhancing ferroptosis. CCK-8 and colony formation were used to detect cell proliferation. Ferroptosis indicator reactive oxygen species (ROS), glutathione (GSH) activity, ATP activity, Fe2+ content, and GPX4 protein expression were detected using ROS assay kit, reduced GSH content assay kit, ATP content assay kit, ferrous ion content assay kit, and western blotting, respectively. The protein levels of HO-1 and Nrf2-central regulators of antioxidant defense and ferroptosis resistance-were assessed by western blot. Tumor changes were observed in nude mice with subcutaneous tumorigenesis. Tamoxifen treatment reduced cell proliferation and colony formation, decreased GSH levels, and downregulated the expression of GPX4, Nrf2, and HO-1. Conversely, it increased ROS fluorescence intensity and Fe2⁺ accumulation and impaired the formation of MCF-7 organoids. Overexpression of Nrf2 reversed the effect of tamoxifen. Compared with MCF-7 treated with tamoxifen, in MCF-7 TAMR treated with tamoxifen, the cell proliferation, clone number, ATP activity, and the mRNA expression of Nrf2 and HO-1 significantly increased. The subcutaneous tumor formation experiment in nude mice confirmed that tumors simultaneously treated with ML385 and tamoxifen can further shrink the tumor. The expression of Nrf2 in clinical breast cancer and recurrent breast cancer tissues was significantly higher than that in paracancerous and primary breast cancer tissues, respectively. ML385, the inhibitor of Nrf2, increased ferroptosis via inhibiting Nrf2/HO-1 pathway to enhance the sensitivity of MCF-7 TAMR to tamoxifen.
Collapse
Affiliation(s)
- Jie Yuan
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China.
- Foshan Municipal Key Laboratory of Precision Oncology, Foshan Fosun Chancheng Hospital, Foshan, China.
| | - Wenjun Huang
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Minxia Lin
- Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
| | - Shishen Sun
- Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
| | - Fei Zhong
- Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
| | - Lei Ye
- Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
| | - Hao Yin
- Foshan Municipal Key Laboratory of Precision Oncology, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Xiaowei Ou
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China
- Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China
| | - Zhiqiang Zeng
- Department of General Surgery, Foshan Fosun Chancheng Hospital, Foshan, China.
- Foshan Clinical Medical School, Guangzhou University of Chinese Medicine, Foshan, China.
| |
Collapse
|
15
|
Sun Y, Ma X, Gong Y, Guo H, Zhou C, Hu Q, Zhou Z, Zhang Y, Liang S, Li G. Inhibition of P2X7R by Hypericin improves Diabetic Cardiac Autonomic Neuropathy through the proteasome- Nrf2 - GPX4 signaling axis. Neurotoxicology 2025:S0161-813X(25)00059-2. [PMID: 40412558 DOI: 10.1016/j.neuro.2025.05.008] [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: 02/09/2025] [Revised: 04/29/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Hypericin (HYP), a primary active compound derived from hypericum perforatum has been studied in the context of diabetes. The purpose of this study is to observe whether HYP can improve diabetic cardiac autonomic neuropathy (DCAN) and its possible mechanism. The current findings suggest that multiple drivers of ferroptosis in DCAN converge on the antioxidant protein nuclear factor erythroid 2-related factor 2(Nrf2). Overactivated P2X7 receptor (P2X7R) increases Nrf2 degradation by increasing proteasome activity through calcium ion accumulation. This work showed that HYP inhibited P2X7R expression, leading to elevated Nrf2 levels, thereby counteracting ferroptosis. This inhibition improves abnormal changes in cardiac function during the pathological process of DCAN in diabetic rats, including heart rate (HR), blood pressure (BP), heart rate variability (HRV), and sympathetic nerve discharge (SND). In summary, HYP enhances Nrf2 protein levels by suppressing P2X7R expression, reducing calcium-induced proteasome activity, and inhibits ferroptosis and inflammation. Thus, HYP alleviated DCAN progression.
Collapse
Affiliation(s)
- Yusen Sun
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Xiaoqian Ma
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yanning Gong
- Second Clinical Medical School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Hongmin Guo
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Congfa Zhou
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Qixing Hu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Zhiying Zhou
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Yuanyuan Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Shangdong Liang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
| | - Guilin Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China.
| |
Collapse
|
16
|
Gong Y, Zhang W, Wang X, Zheng S, Liu H, Lin Q, Wang M, Ma J, Zhang Y, Li T, Hu Y. A previously uncharacterized role of TAp73 in ferroptosis by modulating oxidative homeostasis in cervical cancer. Cancer Lett 2025:217815. [PMID: 40409453 DOI: 10.1016/j.canlet.2025.217815] [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: 04/07/2025] [Revised: 05/17/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
While the tumor-suppressive functions of p53 are well established, the role of its homolog, TAp73, in cancer remains incompletely characterized and is a subject of active investigation. In this study, we observed downregulation of TAp73 protein expression in cervical cancer tissues, which significantly correlated with adverse clinical outcomes. Through co-expression network analysis, we identified functional associations between TAp73 and key pathways involved in lipid metabolism and redox homeostasis-both critical regulators of ferroptosis, an iron-dependent form of programmed cell death mediated by lipid peroxidation. Mechanistically, we demonstrate that TAp73 promotes ferroptosis by directly upregulating the transcription of β-transducin repeat-containing protein (β-TRCP), thereby facilitating the ubiquitin-dependent degradation of nuclear factor erythroid 2-related factor 2 (NRF2), a master regulator of cellular antioxidant defenses. This TAp73-mediated suppression of NRF2 activity renders cells more susceptible to ferroptotic death. Furthermore, TAp73 expression is transcriptionally induced during ferroptosis through the combined inactivation of enhancer of zeste homolog 2 (EZH2), a core component of polycomb repressive complex 2, and activation of E2F transcription factor 1 (E2F1). Notably, pharmacological inhibition of EZH2 synergized with sulfasalazine (SAS) to enhance ferroptosis in vivo, an effect largely dependent on TAp73. Together, these findings delineate a novel ferroptosis regulatory axis-EZH2/TAp73/β-TRCP/NRF2-and highlight its potential as a therapeutic target for cervical cancer intervention.
Collapse
Affiliation(s)
- Yafan Gong
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Wenxin Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Shanliang Zheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Hao Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Qingyu Lin
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Meiqi Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001
| | - Jiangwen Ma
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001
| | - Yi Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Tianyu Li
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, China, 150001; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, China,450000.
| |
Collapse
|
17
|
Falcão AS, Pedro ML, Tenreiro S, Seabra MC. Targeting Lysosomal Dysfunction and Oxidative Stress in Age-Related Macular Degeneration. Antioxidants (Basel) 2025; 14:596. [PMID: 40427478 PMCID: PMC12108406 DOI: 10.3390/antiox14050596] [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: 04/04/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of vision loss in the Western world, and it currently lacks effective therapy. It is believed that AMD initiates in the aged retinal pigment epithelium (RPE), which presents lysosomal dysfunction and oxidative stress (OxS) that ultimately leads to RPE damage and AMD progression. AMD is a complex pathology, so multitarget treatments are required to act on different pathways, presenting several challenges. In this review, we discuss the current knowledge on the pathogenesis of this disease, focusing mainly on lysosomal dysfunction and OxS. Because transcription factors regulate homeostasis, the transcription factor EB (TFEB), which controls lysosomal function and biogenesis, and the nuclear factor erythroid 2-related factor 2 (NRF2), which manages OxS, have been proposed as promising targets for disease intervention. Finally, we discuss the interplay of these pathways for a potential synergistic effect on AMD-targeted therapies, as they could change the course of today's available treatments for AMD.
Collapse
Affiliation(s)
| | | | | | - Miguel C. Seabra
- iNOVA4Health, NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| |
Collapse
|
18
|
Liu L, Che B, Zhang W, Du D, Zhang D, Li J, Chen Z, Yu X, Ye M, Wang W, Li Z, Xie F, Wang Q, Chen L, Shao J. Mechanistic insights into the role of FAT10 in modulating NCOA4-mediated ferroptosis in pancreatic acinar cells during acute pancreatitis. Cell Death Dis 2025; 16:385. [PMID: 40374601 DOI: 10.1038/s41419-025-07715-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 04/25/2025] [Accepted: 05/02/2025] [Indexed: 05/17/2025]
Abstract
Acute pancreatitis (AP) is characterised by inflammation and cell death in pancreatic tissue, with ferroptosis playing a critical role in its pathophysiology by mediating cellular damage and exacerbating inflammation. This study investigated the role of human leukocyte antigen (HLA)-F adjacent transcript 10 (FAT10) in AP, specifically its involvement in ferroptosis within pancreatic acinar cells. We observed that FAT10 expression was significantly elevated in AP tissues, which correlated with increased ferroptosis. Overexpression of FAT10 in pancreatic acinar cells enhances ferroptosis, whereas its knockdown reduced levels of ferroptosis markers. Furthermore, we confirmed that FAT10 enhanced ferroptosis in pancreatic acinar cells primarily by upregulating nuclear receptor coactivator 4 (NCOA4) expression. Mechanistic investigations revealed that FAT10 regulates NCOA4 expression to promote ferroptosis in a complex manner. FAT10 inhibits NCOA4 ubiquitination by reducing ubiquitin-NCOA4 complexes. Meanwhile, NCOA4 expression increased alongside the increase in FAT10-NCOA4 complexes, which are resistant to proteasomal degradation. Notably, we identified silibinin, a natural compound, as an effective inhibitor of the FAT10-NCOA4 axis, leading to reduced ferroptosis and alleviation of pancreatic damage in vivo. Silibinin treatment decreased the levels of ferroptosis-related proteins and inflammatory markers in both cell and animal models. Our findings highlight the FAT10-NCOA4 axis as a crucial regulator of ferroptosis in pancreatic acinar cells and suggest that targeting this pathway could offer a therapeutic strategy for mitigating AP. This study provides new insights into the regulatory mechanisms of ferroptosis in pancreatic acinar cells, identifying FAT10 as a potential therapeutic target for AP management.
Collapse
Affiliation(s)
- Lingpeng Liu
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ben Che
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenming Zhang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dongnian Du
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dandan Zhang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jiajuan Li
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zehao Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuzhe Yu
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Miao Ye
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Wang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zijing Li
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fei Xie
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qing Wang
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Liver Cancer Institute, Nanchang University, Nanchang, China
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Leifeng Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- Precision Oncology Medicine Center, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.
| | - Jianghua Shao
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- Liver Cancer Institute, Nanchang University, Nanchang, China.
- Jiangxi Province Clinical Research Center of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Nanchang University, Nanchang, China.
| |
Collapse
|
19
|
Hasan SK, Jayakumar S, Espina Barroso E, Jha A, Catalano G, Sandur SK, Noguera NI. Molecular Targets of Oxidative Stress: Focus on Nuclear Factor Erythroid 2-Related Factor 2 Function in Leukemia and Other Cancers. Cells 2025; 14:713. [PMID: 40422216 DOI: 10.3390/cells14100713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/04/2025] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that plays a central role in regulating cellular responses to oxidative stress. It governs the expression of a broad range of genes involved in antioxidant defense, detoxification, metabolism, and other cytoprotective pathways. In normal cells, the transient activation of Nrf2 serves as a protective mechanism to maintain redox homeostasis. However, the persistent or aberrant activation of Nrf2 in cancer cells has been implicated in tumor progression, metabolic reprogramming, and resistance to chemotherapy and radiotherapy. These dual roles underscore the complexity of Nrf2 signaling and its potential as a therapeutic target. A deeper understanding of Nrf2 regulation in both normal and malignant contexts is essential for the development of effective Nrf2-targeted therapies. This review provides a comprehensive overview of Nrf2 regulation and function, highlighting its unique features in cancer biology, particularly its role in metabolic adaptation and drug resistance. Special attention is given to the current knowledge of Nrf2's involvement in leukemia and emerging strategies for its therapeutic modulation.
Collapse
Affiliation(s)
- Syed K Hasan
- Hasan Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai 400094, India
| | - Sundarraj Jayakumar
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai 400094, India
| | | | - Anup Jha
- Hasan Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai 400094, India
| | - Gianfranco Catalano
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, 00042 Rome, Italy
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00042 Rome, Italy
| | - Santosh K Sandur
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Department of Life Sciences, Homi Bhabha National Institute, Mumbai 400094, India
| | - Nelida I Noguera
- Santa Lucia Foundation, I.R.C.C.S. Via del Fosso di Fiorano, 00042 Rome, Italy
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00042 Rome, Italy
| |
Collapse
|
20
|
Currais A, Sanchez K, Soriano-Castell D, Dar NJ, Evensen KG, Soriano S, Maher P. Transcriptomic signatures of oxytosis/ferroptosis are enriched in Alzheimer's disease. BMC Biol 2025; 23:132. [PMID: 40369584 PMCID: PMC12080116 DOI: 10.1186/s12915-025-02235-6] [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: 12/04/2024] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Oxytosis/ferroptosis is a form of non-apoptotic regulated cell death characterized by specific changes in the redox balance that lead to lethal lipid peroxidation. It has been hypothesized recently that aging predisposes the brain to the activation of oxytosis/ferroptosis in Alzheimer's disease (AD), and consequently that inhibition of oxytosis/ferroptosis offers a path to develop a new class of therapeutics for the disease. The goal of the present study was to investigate the occurrence of oxytosis/ferroptosis in the AD brain by examining transcriptomic signatures of oxytosis/ferroptosis in cellular and animal models of AD as well as in human AD brain samples. RESULTS Since oxytosis/ferroptosis has been poorly defined at the RNA level, the publicly available datasets are limited. To address this limitation, we developed TrioSig, a gene signature generated from transcriptomic data of human microglia, astrocytes, and neurons treated with inducers of oxytosis/ferroptosis. It is shown that the different signatures of oxytosis/ferroptosis are enriched to varying extents in the brains of AD mice and human AD patients. The TrioSig signature was the most frequently found enriched, and bioinformatic analysis of its composition identified genes involved in the integrated stress response (ISR). It was confirmed in nerve cell culture that oxytosis/ferroptosis induces the ISR via phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) and activating transcription factor 4 (ATF4) signaling. CONCLUSIONS Our data support the involvement of oxytosis/ferroptosis in AD. The implications of the ISR for the progression and prevention of AD are discussed.
Collapse
Affiliation(s)
- Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA, 92037, USA.
| | - Kayla Sanchez
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - David Soriano-Castell
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Nawab John Dar
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - K Garrett Evensen
- The Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Salvador Soriano
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA, 92037, USA.
| |
Collapse
|
21
|
Li X, Sun YX, Tjahjono AW, Wei Y, Li X, Zheng QH, Qi WC, Liang FR. Acupuncture attenuates myocardial ischemia/reperfusion injury-induced ferroptosis via the Nrf2/HO-1 pathway. Chin Med 2025; 20:61. [PMID: 40346679 PMCID: PMC12065278 DOI: 10.1186/s13020-025-01114-0] [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: 02/24/2025] [Accepted: 04/27/2025] [Indexed: 05/11/2025] Open
Abstract
AIMS To observe the effect of electro-acupuncture (EA) on cardiomyocytes ferroptosis induced by myocardial ischemia/reperfusion injury (MIRI) in mice and to investigate whether this effect occurs via the nuclear factor-E2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) signalling pathway. MATERIALS AND METHODS Firstly, Fe2+ in the hearts and serum of mice from both the sham-operated (SO) group and MIRI group was measured to ascertain whether ferroptosis had occurred in the cardiomyocytes of mice in MIRI group. In the second phase, EA was administered, with sham acupuncture (SA) group as the comparator, to investigate the protective effects of EA on ferroptosis in MIRI cardiomyocytes and cardiac function. Additionally, we studied the levels of Nrf2 and HO-1 within the myocardium. In the third phase, Nrf2 inhibitor ML385 and agonist DMF were applied to observe the impact of inhibiting Nrf2 on the therapeutic efficacy of EA. RESULTS Compared with SO group, MIRI group showed increased iron deposition, along with a significant decrease in Nrf2 and HO-1 levels. Compared with MIRI group, MIRI + EA group exhibited significantly improved cardiac function and reduced cardiac iron deposition, accompanied by increased Nrf2 and HO-1 levels. Furthermore, the therapeutic effect of MIRI + EA group was superior to that of MIRI + SA group. Administration of ML385 partially blocked the anti-ferroptotic and cardioprotective effects of EA, while EA treatment exhibited similar effects to dimethyl fumarate (DMF) intervention. CONCLUSION EA alleviates ferroptosis-induced damage in MIRI in mice via the Nrf2/HO-1 pathway, providing modern scientific evidence for the application of acupuncture in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiao Li
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
| | - Yu-Xin Sun
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
| | - Adi Wirawan Tjahjono
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
| | - Ying Wei
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
| | - Xiang Li
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
- Sichuan Provincial Key Laboratory of Acupuncture and Chronobiology, Chengdu, 610075, People's Republic of China
| | - Qian-Hua Zheng
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
- Sichuan Provincial Key Laboratory of Acupuncture and Chronobiology, Chengdu, 610075, People's Republic of China
| | - Wen-Chuan Qi
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China.
- Sichuan Provincial Key Laboratory of Acupuncture and Chronobiology, Chengdu, 610075, People's Republic of China.
| | - Fan-Rong Liang
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China.
- Sichuan Provincial Key Laboratory of Acupuncture and Chronobiology, Chengdu, 610075, People's Republic of China.
| |
Collapse
|
22
|
Chen C, Wang L, Cui XF, Shang XY, Bai SH, Li L, Wang N, Han ZG. SCARA5 deficiency inhibits ferroptosis via regulating iron homeostasis and results in sorafenib resistance in hepatocellular carcinoma. Cell Signal 2025; 129:111656. [PMID: 39954713 DOI: 10.1016/j.cellsig.2025.111656] [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: 10/30/2024] [Revised: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
SCARA5 (Scavenger Receptor Class A Member 5), a member of scavenger receptor class A, is a type II transmembrane protein. Previous studies, including our own, have suggested that SCARA5 acts as a tumor suppressor in various cancers. Additionally, SCARA5 has been identified as a ferritin receptor that facilitates iron delivery independent of transferrin. However, it remains unclear whether ferroptosis is involved in the tumor-suppressive function of SCARA5 in hepatocellular carcinoma (HCC). In this study, we found that SCARA5-deficient cells, including mouse embryonic fibroblasts (MEFs) and HCC cells, exhibited reduced sensitivity to ferroptosis induced by erastin and RSL3. We measured the cell viability, cellular reactive oxygen species (ROS), lipid ROS, malondialdehyde (MDA) and ferrous iron concentration to assess the role of SCARA5 in ferroptosis. Mechanistically, we confirmed that SCARA5 might enhance the intracellular availability of bioactive ferrous iron by promoting autophagic degradation of the major iron storage protein ferritin. Furthermore, we found that SCARA5 deficiency contributed to the resistance of HCC cells to sorafenib, a therapeutic agent for HCC, possibly by inhibiting ferroptosis. Collectively, our study revealed the role of SCARA5 in regulating ferroptosis, providing a profound understanding of sorafenib resistance in HCC systemic therapy.
Collapse
Affiliation(s)
- Cong Chen
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lan Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiao-Fang Cui
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xu-Yang Shang
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shi-Hao Bai
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lin Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Na Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ze-Guang Han
- Key Laboratory of Systems Biomedicine (Ministry of Education), State Key Laboratory of Medical Genomics, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
23
|
Taufani IP, Tasminatun S, Harimurti S, Yang LY, Huang CY, Situmorang JH. Tannic Acid Suppresses Ferroptosis Induced by Iron Salophene Complex in Kidney Cells and Prevents Iron Overload-Induced Liver and Kidney Dysfunction in Rats. Biol Trace Elem Res 2025; 203:2701-2713. [PMID: 39207654 DOI: 10.1007/s12011-024-04360-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Iron toxicity intricately links with ferroptosis, a unique form of cell death, and is significantly influenced by lipid peroxidation. Despite its critical role in various diseases and drug development, the association between iron toxicity and ferroptosis remains relatively unexplored. Accidental iron ingestion has emerged as a growing concern, resulting in a spectrum of symptoms ranging from gastrointestinal discomfort to severe outcomes, including mortality. This research introduces tannic acid (TA), which contains numerous phenol groups, as a powerful antiferroptotic agent. In male Wistar rats, even a modest dose of TA (7.5 mg/kg) significantly curtailed thiobarbituric acid reactive substances (TBARS), a well-established indicator of lipid peroxidation, and mitigated iron accumulation induced by ferrous sulfate (FeSO4) in the liver and kidney. The evidence supporting TA's protective function against iron-triggered liver and kidney dysfunction was substantiated by assessing specifically the levels of blood urea nitrogen (BUN) and alanine aminotransferase (ALT). In cell models using ferroptosis inducers such as iron-salophene (FeSP) and RAS-selective lethal 3 (RSL3), tannic acid (TA) exhibited superior protective capabilities compared to the traditional iron chelator, deferoxamine (DFO). Nrf2 and HO-1, regulators of antioxidant defense genes, are implicated in controlling ferroptosis. The expression of Nrf2 and HO-1 increased with TA treatment in the presence of FeSP, indicating their role in reducing lipid ROS levels. Additionally, TA significantly reduced the heightened levels of COX2, a marker associated with ferroptosis. In summary, the remarkable antiferroptosis activity of TA is likely due to its combined iron-chelating and antioxidant properties. With its safety profile for oral consumption, TA may offer benefits in cases of accidental iron ingestion and conditions like hemochromatosis.
Collapse
Affiliation(s)
- Indra Putra Taufani
- Graduate Institute of Pharmacy, China Medical University, Taichung, Taiwan
- Department of Pharmacist Profession Education, Faculty of Medicine and Health Sciences, Universitas Muhammadiyah Yogyakarta, Yogyakarta, Indonesia
| | - Sri Tasminatun
- School of Pharmacy, Faculty of Medicine and Health Sciences, Universitas Muhammadiyah Yogyakarta, Yogyakarta, Indonesia
| | - Sabtanti Harimurti
- School of Pharmacy, Faculty of Medicine and Health Sciences, Universitas Muhammadiyah Yogyakarta, Yogyakarta, Indonesia
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.
- Laboratory for Neural Repair, China Medical University Hospital, Taichung, Taiwan.
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Buddhist Tzu Chi General, Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.
- Graduate Institute of Medical Science, China Medical University, Taichung, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
- Department of Biotechnology, Asia University, Taichung, Taiwan.
- Center of General Education, Tzu Chi University of Science and Technology, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.
| | - Jiro Hasegawa Situmorang
- Center for Biomedical Research, National Research and Innovation Agency (BRIN), Cibinong, Indonesia.
| |
Collapse
|
24
|
Zhang N, Fan Y, Chen J, Gu J, Yan X. MAPK14 drives Ferroptosis and immune dysfunction in pediatric Sepsis-induced acute lung injury. Cell Immunol 2025; 411-412:104948. [PMID: 40209319 DOI: 10.1016/j.cellimm.2025.104948] [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: 02/15/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
OBJECTIVE Sepsis-induced acute lung injury (ALI) is driven by inflammation, oxidative stress, and immune suppression. MAPK14 (p38α) plays a role in ferroptosis and immune regulation, but its specific function in pediatric sepsis remains unclear. Therefore, our study aimed to explore the role and underlying mechanism of MAPK14 in pediatric sepsis. METHODS Bioinformatics analysis of GSE26440 and FerrDb identified ferroptosis-related genes in pediatric sepsis. STRING database was used to predict the proteins associated with MAPK14. MAPK14 expression in whole blood samples, LPS-treated MLE-12 cells, and a CLP mouse model was detected by qRT-PCR and western blot. Ferroptosis was assessed by measuring MDA, GSH, and Fe2+ levels, while ROS accumulation was analyzed using DCFH-DA staining and DHE staining. A cycloheximide (CHX) assay was performed to assess TTP53 protein stability. MPO immunohistochemistry and PD-L1 immunofluorescence assessed neutrophil infiltration, and flow cytometry evaluated neutrophil apoptosis. RESULTS Bioinformatics analysis of GSE26440 and FerrDb identified MAPK14 as a ferroptosis-related gene in pediatric sepsis. MAPK14 expression was upregulated in sepsis patient samples, LPS-treated MLE-12 cells and CLP mouse lung tissues. Overexpression of MAPK14 led to increased MDA and Fe2+ levels, reduced GSH, and elevated ROS fluorescence intensity, confirming its role in promoting ferroptosis. Mechanistically, MAPK14 upregulated TTP53, which in turn suppressed SLC7A11 and GPX4, further driving ferroptosis. MAPK14 overexpression stabilized TTP53 and enhanced its activity. Additionally, MAPK14 enhanced MPO and PD-L1 expression to promote neutrophil infiltration and immune suppression. Additionally, MAPK14 overexpression inhibited neutrophil apoptosis, promoted neutrophil infiltration and enhanced immune suppression. CONCLUSION MAPK14 drives ferroptosis via the TTP53/SLC7A11/GPX4 pathway and exacerbates immune suppression by promoting neutrophil infiltration.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Surgery Intensive Care Unit (SICU), Children's Hospital of Soochow University, Suzhou, Jiangsu 215008, China
| | - Yuanyuan Fan
- Department of Pediatrics, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, China
| | - Juan Chen
- Nanjing Kangze Medical Testing Co., Ltd., Nanjing, Jiangsu 211100, China
| | - Juan Gu
- Department of Emergency, Yinchuan Maternal and Child Health Hospital, Yinchuan, Ningxia 750000, China
| | - Xiangming Yan
- Department of Urology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215008, China.
| |
Collapse
|
25
|
Wang L, Zhang Y, Yue J, Zhou R. The Role of Ubiquitination on Macrophages in Cardiovascular Diseases and Targeted Treatment. Int J Mol Sci 2025; 26:4260. [PMID: 40362498 PMCID: PMC12072125 DOI: 10.3390/ijms26094260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide, with macrophage dysfunction playing a central role in its pathogenesis. Ubiquitination, a critical post-translational modification, regulates diverse macrophage functions, including lipoprotein metabolism, inflammation, oxidative stress, mitophagy, autophagy, efferocytosis, and programmed cell death (pyroptosis, necroptosis, ferroptosis, and apoptosis). This review highlights the regulatory roles of ubiquitination in macrophage-driven CVD progression, focusing on its effects on cholesterol metabolism, inflammation, activation, polarization, and the survival of macrophages. Targeting ubiquitination pathways has therapeutic potential by enhancing macrophage autophagy, reducing inflammation, and improving plaque stability. However, challenges, such as off-target effects, ubiquitination crosstalk, and macrophage heterogeneity, must be addressed. By integrating advances in ubiquitination biology, therapeutic strategies can be developed to mitigate CVD and other macrophage-driven inflammatory diseases. This review underscores the potential of ubiquitination-targeting therapies for mitigating CVD and highlights the key areas for further investigation.
Collapse
Affiliation(s)
- Li Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianming Yue
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ronghua Zhou
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China; (L.W.); (Y.Z.); (J.Y.)
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
26
|
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
|
27
|
Conjard-Duplany A, Osseni A, Lamboux A, Mouradian S, Picard F, Moncollin V, Angleraux C, Dorel-Dubois T, Puccio H, Leblanc P, Galy B, Balter V, Schaeffer L, Gangloff YG. Muscle mTOR controls iron homeostasis and ferritinophagy via NRF2, HIFs and AKT/PKB signaling pathways. Cell Mol Life Sci 2025; 82:178. [PMID: 40293459 PMCID: PMC12037468 DOI: 10.1007/s00018-025-05695-9] [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: 09/02/2024] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
Balanced mTOR activity and iron levels are crucial for muscle integrity, with evidence suggesting mTOR regulates cellular iron homeostasis. In this study, we investigated iron metabolism in muscle-specific mTOR knockout mice (mTORmKO) and its relation to their myopathy. The mTORmKO mice exhibited distinct iron content patterns across muscle types and ages. Slow-twitch soleus muscles initially showed reduced iron levels in young mice, which increased with the dystrophy progression but remained within control ranges. In contrast, the less affected fast-twitch muscles maintained near-normal iron levels from a young age. Interestingly, both mTORmKO muscle types exhibited iron metabolism markers indicative of iron excess, including decreased transferrin receptor 1 (TFR1) and increased levels of ferritin (FTL) and ferroportin (FPN) proteins. Paradoxically, these changes were accompanied by downregulated Ftl and Fpn mRNA levels, indicating post-transcriptional regulation. This discordant regulation resulted from disruption of key iron metabolism pathways, including NRF2/NFE2L2, HIFs, and AKT/PKB signaling. Mechanistically, mTOR deficiency impaired transcriptional regulation of iron-related genes mediated by NRF2 and HIFs. Furthermore, it triggered ferritin accumulation through two NRF2 mechanisms: (1) derepression of ferritin translation via suppression of the FBXL5-IRP axis, and (2) autophagosomal sequestration driven by NCOA4-dependent ferritin targeting to autophagosomes, coupled with age-related impairments of autophagy linked to chronic AKT/PKB activation. Three-week spermidine supplementation in older mTORmKO mice was associated with normalized AKT/PKB-FOXO signaling, increased endolysosomal FTL and reduced total FTL levels in the dystrophic soleus muscle. These findings underscore mTOR's crucial role in skeletal muscle iron metabolism and suggest spermidine as a potential strategy to address impaired ferritinophagy due to autophagy blockade in dystrophic muscle.
Collapse
Affiliation(s)
- Agnès Conjard-Duplany
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR 5261, 8 avenue Rockefeller, Lyon, 69008, France.
| | - Alexis Osseni
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR 5261, 8 avenue Rockefeller, Lyon, 69008, France
| | - Aline Lamboux
- Laboratoire de Géologie de Lyon: Terre, Planètes, Environnement, UMR 5276, Ecole Normale Supérieure de Lyon, 46, allée d'Italie, Lyon, Cedex 07, 69364, France
| | - Sandrine Mouradian
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR 5261, 8 avenue Rockefeller, Lyon, 69008, France
| | - Flavien Picard
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR 5261, 8 avenue Rockefeller, Lyon, 69008, France
| | - Vincent Moncollin
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR 5261, 8 avenue Rockefeller, Lyon, 69008, France
| | - Céline Angleraux
- Université Claude Bernard Lyon 1, CNRS UAR3444, Inserm US8, ENS de Lyon, AniRA-PBES, SFR Biosciences, Lyon, 69007, France
| | - Tiphaine Dorel-Dubois
- Université Claude Bernard Lyon 1, CNRS UAR3444, Inserm US8, ENS de Lyon, AniRA-PBES, SFR Biosciences, Lyon, 69007, France
| | - Hélène Puccio
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR 5261, 8 avenue Rockefeller, Lyon, 69008, France
| | - Pascal Leblanc
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR 5261, 8 avenue Rockefeller, Lyon, 69008, France
| | - Bruno Galy
- German Cancer Research Center (DKFZ), Division of Virus-associated Carcinogenesis (F170), Heidelberg, Germany
- IB-Cancer Research Foundation, Science Park 2, 66123, Saarbrücken, Germany
| | - Vincent Balter
- Laboratoire de Géologie de Lyon: Terre, Planètes, Environnement, UMR 5276, Ecole Normale Supérieure de Lyon, 46, allée d'Italie, Lyon, Cedex 07, 69364, France
| | - Laurent Schaeffer
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR 5261, 8 avenue Rockefeller, Lyon, 69008, France
- Centre de Biotechnologie Cellulaire, Hospices Civils de Lyon, Lyon, France
| | - Yann-Gaël Gangloff
- Laboratoire Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Institut NeuroMyoGène, Université Claude Bernard Lyon 1, INSERM U1315, CNRS UMR 5261, 8 avenue Rockefeller, Lyon, 69008, France.
| |
Collapse
|
28
|
Jinson S, Zhang Z, Lancaster GI, Murphy AJ, Morgan PK. Iron, lipid peroxidation, and ferroptosis play pathogenic roles in atherosclerosis. Cardiovasc Res 2025; 121:44-61. [PMID: 39739567 DOI: 10.1093/cvr/cvae270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Oxidation of lipids, excessive cell death, and iron deposition are prominent features of human atherosclerotic plaques. While extensive research has established the detrimental roles of lipid oxidation and apoptosis in atherosclerosis development, the involvement of iron in atherogenesis is not yet fully understood. With the emergence of an iron-dependent form of cell death termed ferroptosis, new attention has been brought to the complex inter-play among iron, ferroptosis, and atherosclerosis. Mechanistically, ferroptosis is caused by the lethal accumulation of iron-mediated lipid peroxides. Emerging studies have underscored ferroptosis as a contributor to worsened atherosclerosis. Herein, we review the evidence that oxidative damage and iron overload in the context of atherosclerosis may promote ferroptosis within plaques. Furthermore, we summarize recent findings of lipid peroxidation, thereby potentially ferroptosis, in various plaque cell types-such as endothelial cells, macrophages, dendritic cells, T cells, and vascular smooth muscle cells-across different stages of atherosclerosis. Understanding how these processes influence atherosclerotic plaque progression may permit targeting stage-dependent ferroptosis in each cell population and could provide a rationale for developing cell type-specific intervention strategies to mitigate atherogenic ferroptosis effectively.
Collapse
Affiliation(s)
- Swetha Jinson
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Ziyang Zhang
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Graeme I Lancaster
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Andrew J Murphy
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Pooranee K Morgan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| |
Collapse
|
29
|
Zhang Y, Tang Q, Yao J, Liu H, Xu C, Guo Z, Liu S, Zhao R. Yi-Nao-Jie-Yu Prescription Relieves Post-Stroke Depression by Mitigating Ferroptosis in Hippocampal Neurons Via Activating the Nrf2/GPX4/SLC7A11 Pathway. J Neuroimmune Pharmacol 2025; 20:35. [PMID: 40214929 PMCID: PMC11991945 DOI: 10.1007/s11481-024-10167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 12/14/2024] [Indexed: 04/14/2025]
Abstract
Post-stroke depression (PSD) poses a serious impact on patients' life quality. Effective drugs to treat this annoying disease are still being sought. Yi-nao-jie-yu (YNJY) prescription has been found to relieve PSD; however, the underlying mechanisms remain unelucidated. This work elucidated the therapeutic effects and mechanisms underlying YNJY prescription in PSD. PSD rat model was treated with YNJY prescription and ML385. Depression-like behaviors of rats was appraised. Hematoxylin-eosin, Nissl, and NeuN immunofluorescence staining were performed to observe hippocampal neuronal damage. Transmission electron microscopy was used to assess hippocampal mitochondrial damage. Commercial kits and western blotting were adopted to research ferroptosis-related factors and Nrf2/GPX4/SLC7A11 signals. In vitro experiments were performed using rat hippocampal neurons to explore the mechanism by which YNJY prescription relieves PSD. In PSD rats, YNJY prescription relieved depression-like behaviors, attenuated hippocampal neuronal damage, mitigated hippocampal ferroptosis and mitochondrial damage, and activated hippocampal Nrf2/GPX4/SLC7A11 pathway. By in vitro experiments, erastin treatment exacerbated hippocampal neuronal damage, ferroptosis, mitochondrial damage, and lipid peroxidation; however, YNJY prescription abolished these erastin-induced effects. In the erastin-treated hippocampal neuronal model of PSD, ML385 treatment increased ferroptosis, hippocampal neuronal damage, and lipid peroxidation; however, YNJY prescription counteracted these ML385-induced effects. By in vivo study, ML385 reversed the relief of YNJY prescription on depressive-like behaviors of PSD rats, and the inhibition on ferroptosis in PSD rats' hippocampus. YNJY prescription relieves PSD by blocking ferroptosis via activating the Nrf2/GPX4/SLC7A11 pathway. It may be a promising agent for treating PSD clinically.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Encephalopathy, Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, 101300, China
| | - Qisheng Tang
- Department of Encephalopathy, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jin Yao
- Department of Acupuncture, Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, 101300, China
| | - Hongwei Liu
- Department of Encephalopathy, Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, 101300, China
| | - Changmin Xu
- Department of Encephalopathy, Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, 101300, China
| | - Zechun Guo
- Department of Encephalopathy, Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, 101300, China
| | - Shuqing Liu
- Department of Encephalopathy, Shunyi Hospital, Beijing Hospital of Traditional Chinese Medicine, Beijing, 101300, China
| | - Ruizhen Zhao
- Prevention Center, The Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China.
- , No. 51, Andingmen Outside Xiaoguan Street, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
30
|
Cai D, Li J, Peng Z, Fu R, Chen C, Liu F, Li Y, Su Y, Li C, Chen W. Interplay of Ferroptosis, Cuproptosis, Autophagy and Pyroptosis in Male Infertility: Molecular Crossroads and Therapeutic Opportunities. Int J Mol Sci 2025; 26:3496. [PMID: 40331931 PMCID: PMC12026609 DOI: 10.3390/ijms26083496] [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: 02/08/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Male infertility is intricately linked to dysregulated cell death pathways, including ferroptosis, cuproptosis, pyroptosis, and autophagy. Ferroptosis, driven by iron-dependent lipid peroxidation through the Fenton reaction and inactivation of the GPX4/Nrf2/SLC7A11 axis, disrupts spermatogenesis under conditions of oxidative stress, environmental toxin exposure, or metabolic disorders. Similarly, cuproptosis-characterized by mitochondrial dysfunction and disulfide stress due to copper overload-exacerbates germ cell apoptosis via FDX1 activation and NADPH depletion. Pyroptosis, mediated by the NLRP3 inflammasome and gasdermin D, amplifies testicular inflammation and germ cell loss via IL-1β/IL-18 release, particularly in response to environmental insults. Autophagy maintains testicular homeostasis by clearing damaged organelles and proteins; however, its dysregulation impairs sperm maturation and compromises blood-testis barrier integrity. These pathways intersect through shared regulators; reactive oxygen species and mTOR modulate the autophagy-pyroptosis balance, while Nrf2 and FDX1 bridge ferroptosis-cuproptosis crosstalk. Therapeutic interventions targeting these mechanisms have shown promise in preclinical models. However, challenges persist, including the tissue-specific roles of gasdermin isoforms, off-target effects of pharmacological inhibitors, and transgenerational epigenetic impacts of environmental toxins. This review synthesizes current molecular insights into the cell death pathways implicated in male infertility, emphasizing their interplay and translational potential for restoring spermatogenic function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Wei Chen
- Health Science Center, Hunan Normal University, Changsha 410013, China; (D.C.); (J.L.); (Z.P.); (R.F.); (C.C.); (F.L.); (Y.L.); (Y.S.); (C.L.)
| |
Collapse
|
31
|
Jiang Y, Li J, Wang T, Gu X, Li X, Liu Z, Yue W, Li M. VIPAS39 confers ferroptosis resistance in epithelial ovarian cancer through exporting ACSL4. EBioMedicine 2025; 114:105646. [PMID: 40088627 PMCID: PMC11957506 DOI: 10.1016/j.ebiom.2025.105646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/11/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The high mortality rate associated with epithelial ovarian cancer (EOC) is primarily due to recurrence and chemoresistance, underscoring the urgent need for innovative therapeutic approaches that leverage newly identified vulnerabilities in cancer cells. While conventional chemotherapies induce apoptosis by targeting DNA or mitotic machinery, ferroptosis represents a new distinct form of programmed cell death characterised by the accumulation of lipid peroxides. METHODS The sensitivity of different EOC cell lines to ferroptosis inducers was evaluated using cell viability assays and lipid peroxidation measurements. Live-cell imaging with the pH-sensitive CD63-pHuji reporter was performed to track the extracellular export of acyl-CoA synthetase long-chain family member 4 (ACSL4) via exosomes. The upstream regulator of ACSL4 were identified through immunoprecipitation-mass spectrometry (IP-MS) and validated using protein binding assays. Finally, cell-derived xenograft (CDX) and patient-derived xenograft (PDX) models were utilised to evaluate the therapeutic potential overcoming ferroptosis resistance. FINDINGS In this study, we found that interferon (IFN)-γ combined with arachidonic acid (AA), which are endogenous ferroptosis inducers, could initiate ferroptosis in most EOC cells. However, some EOC cells displayed significant resistance. Contrary to the typical increase in ACSL4 protein observed in ferroptosis-sensitive cells, resistant EOC cells exhibited surprisingly low levels of this pro-ferroptotic lipid metabolic protein. Intriguingly, this reduction is attributed to the exosomal expulsion of ACSL4 protein, revealing a distinct cellular mechanism to evade ferroptosis. We further identified VIPAS39 as a pivotal regulator in sorting ACSL4 into late endosomes, thereby facilitating their subsequent release as exosomes. Notably, targeting VIPAS39 not only overcomes the resistance to ferroptotic cell death but also markedly suppresses tumour growth. INTERPRETATION Our findings uncover the crucial role of VIPAS39 in ferroptosis evasion by facilitating the exporting of ACSL4 protein via exosomes, highlighting VIPAS39 as a promising target for ferroptosis-based anti-cancer therapy. FUNDING Funded by Beijing Municipal Natural Science Foundation (Key program Z220011), National Natural Science Foundation of China (NSFC) (T2225006, T2488301, 82272948), Peking University Medicine Youth Science and Technology Innovation 'Sail Plan' Project Type B Medical Interdisciplinary Seed Fund (71006Y3171), GuangDong Basic and Applied Basic Research Foundation (2021A1515110820), and the special fund of the National Clinical Key Speciality Construction Program, P. R. China (2023).
Collapse
Affiliation(s)
- Yuening Jiang
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Jie Li
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Tianzhen Wang
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Xiaoyang Gu
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Xinyu Li
- Department of Animal Science, College of Animal Science, Hebei North University, Zhangjiakou, Hebei Province, China; Department of Gynecology and Obstetrics, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhaofei Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Wei Yue
- Interdisciplinary Eye Research Institute (EYE-X Institute) Bengbu Medical University, Bengbu, Anhui, 233030, China.
| | - Mo Li
- State Key Laboratory of Female Fertility Promotion, Centre for Reproductive Medicine, Department of Obstetrics and Gynaecology, Peking University Third Hospital, Beijing, 100191, China; National Clinical Research Centre for Obstetrics and Gynaecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China; Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
32
|
Xu WX, Wen X, Fu YT, Yang J, Cui H, Fan RF. Nuclear receptor coactive 4-mediated ferritinophagy: a key role of heavy metals toxicity. Arch Toxicol 2025; 99:1257-1270. [PMID: 39928088 DOI: 10.1007/s00204-025-03963-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/15/2025] [Indexed: 02/11/2025]
Abstract
Nuclear receptor coactive 4 (NCOA4) is a specific receptor for ferritinophagy, transporting ferritin to lysosomal degradation, releasing free iron, and excessive iron levels may lead to cellular redox imbalance, contributing to cell death, predominantly ferroptosis. NCOA4 is regulated by a variety of transcriptional, post-transcriptional, translational, and post-translational modifications. Targeted modulation of NCOA4-mediated ferritinophagy has been successfully used as a therapeutic strategy in several disease models. Recent evidences have elucidated that ferritinophagy and ferroptosis played a major role in heavy metals toxicity. In this review, we explored the regulatory mechanism of NCOA4 as the sole receptor for ferritinophagy from multiple perspectives based on previous studies. The significant role of ferritinophagy-mediated ferroptosis in heavy metals toxicity was discussed in detail, emphasizing the great potential of NCOA4 as a target for heavy metals toxicity.
Collapse
Affiliation(s)
- Wan-Xue Xu
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Xue Wen
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Yi-Tong Fu
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Jie Yang
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Han Cui
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China
| | - Rui-Feng Fan
- College of Veterinary Medicine, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China.
- Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an, 271017, Shandong, China.
| |
Collapse
|
33
|
Feng C, Zhang L, Zhou X, Lu S, Guo R, Song C, Zhang X. Redox imbalance drives magnetic property and function changes in mice. Redox Biol 2025; 81:103561. [PMID: 40020452 PMCID: PMC11910372 DOI: 10.1016/j.redox.2025.103561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
The magnetic properties of substances directly determine their response to an externally applied magnetic field, which are closely associated with magnetoreception, magnetic resonance imaging (MRI), and magnetic bioeffects. However, people's understanding of the magnetic properties of living organisms remains limited. In this study, we utilized NRF2 (nuclear factor erythroid 2-related factor 2) deficient mice to investigate the contribution of redox (oxidation-reduction) homeostasis, in which the key process is the transfer of electron, a direct target of magnetic field and origin of paramagnetism. Our results show that the NRF2-/- mice exhibit significantly altered systemic redox state, accompanied by increased magnetic susceptibility, particularly in the liver and spleen. Further analyses reveal that the levels of paramagnetic reactive oxygen species (ROS) in these tissues are markedly elevated compared to wild-type mice. Moreover, the concentrations of Fe2+ and Fe3+ are significantly elevated in NRF2-/- mice, which are directly correlated with the increased magnetic susceptibility. The disrupted redox balance in NRF2-/- mice not only exacerbates oxidative stress and iron deposition, but also induces impairment to the liver and spleen. The findings highlight the combined effects of ROS and iron metabolism in driving magnetic susceptibility changes, providing valuable theoretical insights for further research into magnetic bioeffects and organ-specific sensitivity to magnetic fields.
Collapse
Affiliation(s)
- Chuanlin Feng
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China
| | - Lei Zhang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Xiaoyuan Zhou
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230039, China
| | - Shiyu Lu
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China
| | - Ruowen Guo
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China
| | - Chao Song
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
| | - Xin Zhang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China; Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230039, China.
| |
Collapse
|
34
|
Cuadrado A, Cazalla E, Bach A, Bathish B, Naidu SD, DeNicola GM, Dinkova-Kostova AT, Fernández-Ginés R, Grochot-Przeczek A, Hayes JD, Kensler TW, León R, Liby KT, López MG, Manda G, Shivakumar AK, Hakomäki H, Moerland JA, Motohashi H, Rojo AI, Sykiotis GP, Taguchi K, Valverde ÁM, Yamamoto M, Levonen AL. Health position paper and redox perspectives - Bench to bedside transition for pharmacological regulation of NRF2 in noncommunicable diseases. Redox Biol 2025; 81:103569. [PMID: 40059038 PMCID: PMC11970334 DOI: 10.1016/j.redox.2025.103569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-activated transcription factor regulating cellular defense against oxidative stress, thereby playing a pivotal role in maintaining cellular homeostasis. Its dysregulation is implicated in the progression of a wide array of human diseases, making NRF2 a compelling target for therapeutic interventions. However, challenges persist in drug discovery and safe targeting of NRF2, as unresolved questions remain especially regarding its context-specific role in diseases and off-target effects. This comprehensive review discusses the dualistic role of NRF2 in disease pathophysiology, covering its protective and/or destructive roles in autoimmune, respiratory, cardiovascular, and metabolic diseases, as well as diseases of the digestive system and cancer. Additionally, we also review the development of drugs that either activate or inhibit NRF2, discuss main barriers in translating NRF2-based therapies from bench to bedside, and consider the ways to monitor NRF2 activation in vivo.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eduardo Cazalla
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Raquel Fernández-Ginés
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28007, Madrid, Spain
| | - Karen T Liby
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Manuela G López
- Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain; Instituto Teófilo Hernando, Madrid, Spain
| | - Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jessica A Moerland
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Graduate School of Medicine Tohoku University, Sendai, Japan; Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Keiko Taguchi
- Laboratory of Food Chemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan; Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Sols-Morreale" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
35
|
Zhang Y, Chen S, Chen G, Zhou L, Zhou G, Yu X, Yuan L, Deng W, Wang Z, Li J, Tu Y, Zhang D, li Y, Sammad A, Zhu X, Yin K. The Type III Secretion System (T3SS) of Escherichia Coli Promotes Atherosclerosis in Type 2 Diabetes Mellitus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413296. [PMID: 39807021 PMCID: PMC12005784 DOI: 10.1002/advs.202413296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Large-scale studies indicate a strong relationship between the gut microbiome, type 2 diabetes mellitus (T2DM), and atherosclerotic cardiovascular disease (ASCVD). Here, a higher abundance of the type III secretion system (T3SS) virulence factors of Enterobacteriaceae/Escherichia-Shigella in patients with T2DM-related-ASCVD, which correlates with their atherosclerotic stenosis is reported. Overexpression of T3SS via Citrobacter rodentium (CR) infection in Apoe-/- T2DM mice exacerbated atherosclerotic lesion formation and increased gut permeability. Non-targeted metabolomic and proteomic analysis of mouse serum showed that T3SS caused abnormal glycerophospholipid metabolism in mice. Proteomics, RNA sequencing, and functional analyses showed that T3SS induced ferroptosis in intestinal epithelial cells, partly due to increased expression of ferritin heavy chains (FTH1). This findings first demonstrated that T3SS increases ferroptosis in intestinal epithelial cells, via disrupting the intestinal barrier and upregulation of phosphatidylcholine, thereby exacerbating T2DM-related ASCVD.
Collapse
Affiliation(s)
- Yao‐Yuan Zhang
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510515China
| | - Song‐Tao Chen
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Gang Chen
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Le Zhou
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541199China
| | - Guo‐Liang Zhou
- Department of CardiologyThe Second Affiliated Hospital of Guilin Medical UniversityGuilin541199China
| | - Xin‐Yuan Yu
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Long Yuan
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Wei‐Qian Deng
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Zhen‐Bo Wang
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Jing Li
- Department of Imaging DiagnosisZhujiang Hospital of Southern Medical UniversityGuangzhou510515China
| | - Yi‐Fu Tu
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Da‐Wei Zhang
- Group on the Molecular and Cell Biology of Lipids and Department of PediatricsFaculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaT6G 2R3Canada
| | - Yuan li
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangxi Key Laboratory of Diabetic Systems MedicineGuilin Medical UniversityGuilin541199China
| | - Abdul Sammad
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
| | - Xiao Zhu
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510515China
- Guangzhou Key Laboratory of Metabolic remodeling and Precise Prevention and Control of DiabetesGuangzhou510515China
| | - Kai Yin
- Department of General PracticeThe Fifth Affiliated Hospital of Southern Medical UniversityGuangzhou510515China
- Guangdong Provincial Key Laboratory of Drug Non‐Clinical Evaluation and ResearchGuangzhou510515China
- Guangzhou Key Laboratory of Metabolic remodeling and Precise Prevention and Control of DiabetesGuangzhou510515China
- Guangxi Clinical Research Center for Diabetes and Metabolic DiseasesThe Second Affiliated Hospital of Guilin Medical UniversityGuilin541199China
| |
Collapse
|
36
|
Ye F, Xu Y, Zhu X, Ding Q, Wang Y, Lu S, Chen Y. The mechanism of E3 ubiquitin ligase HERC1 regulating ferroptosis in lung adenocarcinoma cells. Cancer Genet 2025; 292-293:92-99. [PMID: 39983667 DOI: 10.1016/j.cancergen.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/08/2025] [Accepted: 02/01/2025] [Indexed: 02/23/2025]
Abstract
OBJECTIVE Lung adenocarcinoma (LUAD) is the most prevalent subtype of lung cancer. Herein, we probed into the role of E3 ubiquitin protein ligase family member 1 (HERC1) in promoting ferroptosis and inhibiting LUAD cell proliferation by regulating RAF proto-oncogene serine/threonine-protein kinase (C-RAF). METHODS In cultured human normal lung epithelial cells and non-small cell lung adenocarcinoma cell lines, HERC1 expression was determined by RT-qPCR and Western blot tests. PC-9 and Calu-3 cells were transfected with oe-HERC1, oe-C-RAF or their negative controls. Reactive oxygen species (ROS), malondialdehyde (MDA), glutathione (GSH), and Fe2+ levels were assessed by biochemical assays. Cell viability, death, and proliferation were evaluated by CCK-8, LDH and colony formation assays, followed by assessments of HERC1-C-RAF interaction, C-RAF ubiquitin level, and C-RAF protein stability. RESULTS HERC1 was poorly expressed in LUAD cells. HERC1 promoted LUAD cell ferroptosis and repressed their proliferation and migration, corresponding to reduced levels of system xc-, GPX4, and GSH, as well as elevated levels of ROS, MDA, Fe2+, and ACSL4. LUAD cells overexpressing HERC1 displayed decreased C-RAF protein level, HERC1-C-RAF interaction, elevated C-RAF ubiquitin level, and accelerated C-RAF protein degradation, indicating that HERC1 facilitated C-RAF ubiquitin degradation and attenuated C-RAF protein stability via interaction with C-RAF. C-RAF overexpression partially abrogated the regulatory impact of HERC1 on LUAD cell ferroptosis and proliferation. CONCLUSION HERC1 expedites C-RAF ubiquitin degradation by interacting with C-RAF, which consequently promotes ferroptosis, thereby inhibiting LUAD cell proliferation.
Collapse
Affiliation(s)
- Fei Ye
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yi Xu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xujuan Zhu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Qifeng Ding
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Yifei Wang
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China
| | - Songhua Lu
- Haian People's Hospital Department of Thoracic Surgery, Nantong, 226000, Jiangsu, China
| | - Yongbing Chen
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
37
|
Liu X, Wang W, Nie Q, Liu X, Sun L, Ma Q, Zhang J, Wei Y. The Role and Mechanisms of Ubiquitin-Proteasome System-Mediated Ferroptosis in Neurological Disorders. Neurosci Bull 2025; 41:691-706. [PMID: 39775589 PMCID: PMC11979074 DOI: 10.1007/s12264-024-01343-7] [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: 06/24/2024] [Accepted: 09/29/2024] [Indexed: 01/11/2025] Open
Abstract
Ferroptosis is a form of cell death elicited by an imbalance in intracellular iron concentrations, leading to enhanced lipid peroxidation. In neurological disorders, both oxidative stress and mitochondrial damage can contribute to ferroptosis, resulting in nerve cell dysfunction and death. The ubiquitin-proteasome system (UPS) refers to a cellular pathway in which specific proteins are tagged with ubiquitin for recognition and degradation by the proteasome. In neurological conditions, the UPS plays a significant role in regulating ferroptosis. In this review, we outline how the UPS regulates iron metabolism, ferroptosis, and their interplay in neurological diseases. In addition, we discuss the future application of small-molecule inhibitors and identify potential drug targets. Further investigation into the mechanisms of UPS-mediated ferroptosis will provide novel insights and strategies for therapeutic interventions and clinical applications in neurological diseases.
Collapse
Affiliation(s)
- Xin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Wei Wang
- Cancer Biology Institute, Baotou Medical College, Baotou, 014010, China
| | - Qiucheng Nie
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xinjing Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Lili Sun
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Qiang Ma
- Cancer Biology Institute, Baotou Medical College, Baotou, 014010, China
| | - Jie Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| | - Yiju Wei
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- School of Life Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
38
|
Li Z, Liu J, Ju J, Peng X, Zhao W, Ren J, Jia X, Wang J, Gao F. Cordycepin ameliorates morphine tolerance by inhibiting spinal cord ferroptosis and inflammation via targeting SIRT1. Int J Med Sci 2025; 22:2059-2074. [PMID: 40303502 PMCID: PMC12035829 DOI: 10.7150/ijms.108518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
Morphine tolerance caused by long-term use of morphine is a major medical problem. Neuroinflammation plays an important role in morphine tolerance, and currently no drugs have been found for clinical use to alleviate neuroinflammation during morphine tolerance. Cordycepin is the main active component of fungus cordycepin militaris, has been demonstrated to have anti-oxidative stress and anti-inflammatory properties in various diseases. In this study, we established a rat model of morphine tolerance, examined the effect of cordycepin on the development of morphine tolerance, and evaluated its potential regulatory mechanisms. We found that cordycepin treatment ameliorated the development of morphine tolerance, improved mitochondrial damage associated with ferroptosis, by reducing the levels of reactive oxygen species (ROS), malondialdehyde (MDA) and Fe2+, increasing superoxide dismutase (SOD) and glutathione (GSH) levels, and decreasing the secretion of pro-inflammatory factors (IL-1β, IL-6, and TNF-α). Besides, cordycepin upregulated the expression of SIRT1, SLC7A11 and GPX4. Further research found that the above effects of cordycepin on morphine-tolerant rats were abolished by SIRT1 selective inhibitor EX-527. Thus, these findings indicated that cordycepin could ameliorate the development of morphine tolerance by inhibiting spinal cord ferroptosis and inflammation via targeting SIRT1. Collectively, these results demonstrated the protective effects of cordycepin and highlighted its therapeutic potential as a drug component for morphine tolerance treatment and prevention.
Collapse
Affiliation(s)
- Zheng Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
- Department of Anesthesiology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Jie Liu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jie Ju
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaoling Peng
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Wei Zhao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jihao Ren
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Xiaoqian Jia
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jihong Wang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Feng Gao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| |
Collapse
|
39
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [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: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
40
|
Xu Y, Qian X, Cai G, Lin Z, Huang W, Wang C, Wu H, Zhang Y, Sun J, Zhang Q. WTX-L/β-arrestin2/LCN2 axis controls vulnerability to ferroptosis in gastric cancer. iScience 2025; 28:111964. [PMID: 40109379 PMCID: PMC11919608 DOI: 10.1016/j.isci.2025.111964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/20/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
Gastric cancer (GC) is one of the most prevalent and lethal cancers worldwide. Ferroptosis is a form of iron-dependent regulated cell death emerging as a promising strategy for cancer therapy, whereas the regulation mechanism remains unclear. WTX has been recognized as a potential tumor suppressor, but attempts at targeted therapy have not achieved substantial progress. Further research into the structure, function, and mechanisms is urgently needed. Herein, we identified a long isoform of WTX (WTX-L) as a potent ferroptosis effector in GC. Mechanistically, WTX-L competitively interacts with β-arrestin2, disrupting its direct binding to IκBα and subsequently activating the NF-κB/LCN2 pathway. LCN2 further triggers ferroptosis by significantly increasing the labile Fe2+ pool and promoting excessive lipid peroxidation. Blockade of the WTX-L/β-arrestin2/NF-κB/LCN2 axis significantly diminished the activity of ferroptosis inducers (erastin and RSL3) in vivo. Collectively, these findings reveal that targeting the ferroptosis vulnerabilities through WTX-L may represent a promising strategy for GC.
Collapse
Affiliation(s)
- Yangwei Xu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Xuexia Qian
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Department of Pathology, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, Shanxi 710032, China
| | - Guixing Cai
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
- Department of Orthopedic Oncology, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Zhihao Lin
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Weiye Huang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Chuangyuan Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Hongmei Wu
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Yiqiong Zhang
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jingbo Sun
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
41
|
Yan N, Li G, Zhao L, Guo Q, Yang J, Liu J, Zhou W, Gao Y, Luo Y. Crocin promotes ferroptosis in gastric cancer via the Nrf2/GGTLC2 pathway. Front Pharmacol 2025; 16:1527481. [PMID: 40191433 PMCID: PMC11968662 DOI: 10.3389/fphar.2025.1527481] [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: 11/13/2024] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction: Gastric cancer (GC) is characterized by high incidence and poor survival rates. Crocin, a natural carotenoid from saffron, exhibits antioxidant, anti-inflammatory, and anti-tumor properties. Ferroptosis, an iron-dependent cell death driven by lipid peroxidation, plays a critical role in cancer progression and is a potential therapeutic target. This study investigates whether crocin inhibits GC cell proliferation by inducing ferroptosis and explores its underlying mechanisms. Methods: This study employed in vivo and in vitro models to assess crocin's effects on GC cell proliferation, apoptosis, migration, invasion, and ferroptosis. Pathway enrichment analysis was performed on differentially expressed genes post-crocin treatment. Lentiviral vectors were used to knockdown and overexpress GGTLC2, exploring its role in GC progression and crocin's therapeutic effects. The UCSC and JASPAR databases predicted Nrf2 binding sites in the GGTLC2 promoter. Molecular docking evaluated crocin's affinity for Nrf2 and GGTLC2. Immunofluorescence and nuclear-cytoplasmic fractionation assays analyzed Nrf2 expression and localization. ChIP-qPCR determined Nrf2's regulatory role on GGTLC2 and crocin's modulatory effects. Results: The results demonstrated that crocin significantly inhibited the proliferation, migration, and invasion of GC cells while promoting apoptosis. Differentially expressed genes following crocin treatment were predominantly enriched in pathways associated with oxidative stress and ferroptosis. Crocin downregulated the oncogene GGTLC2, thereby suppressing GC cell proliferation, invasion, and migration, while simultaneously promoting apoptosis and ferroptosis. Molecular docking analysis revealed a stable binding affinity between crocin and GGTLC2, suggesting that crocin may directly target GGTLC2 to modulate its expression. Additionally, crocin facilitated the translocation of Nrf2 from the nucleus to the cytoplasm. ChIP-qPCR results confirmed that Nrf2 directly binds to the GGTLC2 promoter region to regulate its expression, and crocin attenuated this binding interaction. Discussion: In conclusion, our findings suggest that crocin, as a promising natural compound for GC therapy, may inhibit ferroptosis in GC cells through the Nrf2/GGTLC2 signaling pathway, thereby suppressing tumor initiation and progression. This study provides novel insights into the molecular mechanisms underlying the anti-tumor effects of crocin and highlights its potential as a therapeutic agent for GC.
Collapse
Affiliation(s)
- Nan Yan
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of High Altitude Medicine in Qinghai Province, Qinghai Province Plateau Medicine Applied and Basic Research Key Laboratory (Qinghai-Utah Plateau Medicine Joint Key Laboratory), Qinghai University, Xining, China
| | - Gaofu Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Linglin Zhao
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of High Altitude Medicine in Qinghai Province, Qinghai Province Plateau Medicine Applied and Basic Research Key Laboratory (Qinghai-Utah Plateau Medicine Joint Key Laboratory), Qinghai University, Xining, China
| | - Qijing Guo
- Department of oncology, Air force medical center. PLA, Beijing, China
| | - Jie Yang
- Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| | - Jianhong Liu
- College of Humanities and Technology, QingHai Open University, Xining, China
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yushuang Luo
- Research Center for High Altitude Medicine, Key Laboratory of High Altitude Medicine (Ministry of Education), Key Laboratory of High Altitude Medicine in Qinghai Province, Qinghai Province Plateau Medicine Applied and Basic Research Key Laboratory (Qinghai-Utah Plateau Medicine Joint Key Laboratory), Qinghai University, Xining, China
- Affiliated Hospital of Qinghai University, Xining, Qinghai, China
| |
Collapse
|
42
|
Ma Y, Wang X, Li Y, Zhao J, Zhou X, Wang X. Mechanisms Associated with Mitophagy and Ferroptosis in Cerebral Ischemia-reperfusion Injury. J Integr Neurosci 2025; 24:26458. [PMID: 40152564 DOI: 10.31083/jin26458] [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: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 03/29/2025] Open
Abstract
Ischemic stroke (IS) constitutes a major threat to human health. Vascular recanalization by intravenous thrombolysis and mechanical thrombolysis remain the most significant and effective methods for relief of ischemia. Key elements of these treatments include achieving blood-vessel recanalization, restoring brain-tissue reperfusion, and preserving the ischemic penumbra. However, in achieving the therapeutic goals of vascular recanalization, secondary damage to brain tissue from cerebral ischemia-reperfusion injury (CIRI) must also be addressed. Despite advancements in understanding the pathological processes associated with CIRI, effective interventions to prevent its onset and progression are still lacking. Recent research has indicated that mitophagy and ferroptosis are critical mechanisms in the development of CIRI, and significantly contribute to the onset and progression of IS and CIRI because of common targets and co-occurrence mechanisms. Therefore, exploring and summarizing the potential connections between mitophagy and ferroptosis during CIRI is crucial. In the present review, we mainly focused on the mechanisms of mitochondrial autophagy and ferroptosis, and their interaction, in the development of CIRI. We believe that the data show a strong relationship between mitochondrial autophagy and ferroptosis with interactive regulation. This information may underpin new potential approaches for the prevention and treatment of IS and subsequent CIRI.
Collapse
Affiliation(s)
- Yugang Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250014 Jinan, Shandong, China
| | - Xuebin Wang
- Postdoctoral Research Station, Shandong University of Traditional Chinese Medicine, 250014 Jinan, Shandong, China
- Department of Neurology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250001 Jinan, Shandong, China
| | - Yahui Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250014 Jinan, Shandong, China
- Department of Gerontology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250001 Jinan, Shandong, China
| | - Jing Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250014 Jinan, Shandong, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, 250399 Jinan, Shandong, China
| | - Xue Zhou
- Postdoctoral Research Station, Shandong University of Traditional Chinese Medicine, 250014 Jinan, Shandong, China
- Division of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014 Jinan, Shandong, China
| | - Xingchen Wang
- Department of Neurology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250001 Jinan, Shandong, China
- The Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250001 Jinan, Shandong, China
| |
Collapse
|
43
|
Sun L, Niu Y, Liao B, Liu L, Peng Y, Li K, Chen X, Chen Q, Bai D. CUR-PDT induces ferroptosis of RA-FLS via the Nrf2/xCT/GPX4 pathway to inhibit proliferation in rheumatoid arthritis. Inflamm Res 2025; 74:53. [PMID: 40085199 DOI: 10.1007/s00011-025-02019-2] [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: 10/11/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVE Ferroptosis is a non-apoptotic cell death mechanism driven by reactive oxygen species (ROS) and iron. Its significance in inflammatory arthritis is well-established, but its role in rheumatoid arthritis (RA) remains uncertain. This study aimed to clarify the mechanisms through which curcumin-mediated photodynamic therapy (CUR-PDT) triggers ferroptosis in RA fibroblast-like synoviocytes (FLSs). METHODS In vivo studies using a collagen-induced arthritis (CIA) rat model evaluated CUR-PDT effects on joint edema, synovial inflammation, and fibrosis through paw volume measurements and H&E and Masson's trichrome staining. The expression of Nrf2, xCT, and GPX4 in FLSs was assessed via ELISA and immunohistochemistry. In vitro, MH7A cells treated with TNF-α were analyzed for viability, proliferation, invasion, and migration through various assays. Mitochondrial potential and morphology were examined using JC-1 staining and transmission electron microscopy (TEM). Ferroptosis biomarkers, including ROS, malondialdehyde (MDA), glutathione (GSH), superoxide dismutase (SOD), and Fe2+ levels, were measured. Nrf2, xCT, and GPX4 levels were quantified with RT-qPCR, Western blot, and immunofluorescence. Small interfering RNA (siRNA) was employed to knock down Nrf2 to validate the effect of CUR-PDT on ferroptosis in RA-FLS. RESULTS The CUR-PDT therapy markedly reduced joint inflammation and collagen deposition in the synovial tissue of CIA rats. It effectively alleviated both inflammation and hyperplasia. Moreover, this therapy facilitated ferroptosis within the synovial tissue. In vitro analyses indicated that CUR-PDT diminished the proliferation and viability of FLSs, resulting in increased ROS levels in the cells. This cascade initiated ferroptosis, as evidenced by decreased glutathione, heightened iron concentrations, mitochondrial shrinkage, and reduced mitochondrial membrane potential. Crucially, the expression of xCT and GPX4 was significantly lowered. Interestingly, knocking down the Nrf2 gene amplified this effect, leading to an even greater reduction in xCT and GPX4 expression. In this context, RA-FLSs exhibited more pronounced ferroptotic traits, including diminished proliferation, invasion, and migration. CONCLUSIONS This study elucidated a mechanism by which CUR-PDT triggers ferroptosis in FLSs through the downregulation of the Nrf2-xCT-GPX4 signaling cascade, thereby effectively hindering the progression of RA and emphasizing the importance of targeting Nrf2 in disease advancement.
Collapse
Affiliation(s)
- Lihua Sun
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yajuan Niu
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Bo Liao
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Linlin Liu
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yi Peng
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Kaiting Li
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xinhua Chen
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qing Chen
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Dingqun Bai
- Department of Rehabilitation Medicine, Key Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health Commission, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
44
|
Zhang H, Zhang J, Jing X, Huang K, Chen Y, Shen Q, Tao E, Lin D. Involvement of the STAT3/HIF-1α signaling pathway in α-synuclein-induced ferroptosis. Biochem Biophys Res Commun 2025; 752:151419. [PMID: 39946981 DOI: 10.1016/j.bbrc.2025.151419] [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: 08/21/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/24/2025]
Abstract
Oligomeric α-synuclein (α-syn) aggregates, which are a critical pathological feature of Parkinson's disease (PD), can induce neuroinflammation and neurodegeneration. Our previous study revealed a decrease in IL6ST/JAK2/STAT3/HIF-1α pathway in α-syn-induced microglia. As we all know, the JAK2/STAT3 signaling pathway is essential for modulating inflammation, controlling cell growth and exhibiting antiapoptotic responses. However, the precise role of STAT3/HIF-1α in the ferroptosis of α-syn pathology has not been identified in vivo. In this study, above all, we successfully established α-syn-induced mouse models of Parkinson's disease. Our immunohistochemistry results demonstrated that α-syn could activate IL6ST/STAT3/HIF-1α pathway in a model of α-syn-induced PD. We further conducted transcriptomic analysis on a mouse model of α-syn-induced PD, and GSEA revealed an association with ferroptosis. Consequently, we focused on investigating how α-syn might regulate the transcriptional activation of HSPB1. In conclusion, we determined the relationship between ferroptosis and the STAT3/HIF-1α pathway in α-syn-related pathology in vivo. Oligomeric α-syn could induce ferroptosis via the STAT3/HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Han Zhang
- Department of Neurology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Department of Neurology, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Jieli Zhang
- Department of Neurology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xiuna Jing
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kaixun Huang
- Department of Neurology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Ying Chen
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qingyu Shen
- Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Enxiang Tao
- Department of Neurology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China; Department of Neurology, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine of Sun Yat-sen University, Guangzhou, China.
| | - Danyu Lin
- Department of Neurology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
45
|
Zhang Y, Hu J, Zhang Y, Ci X. Amentoflavone protects against cisplatin-induced acute kidney injury by modulating Nrf2-mediated oxidative stress and ferroptosis and partially by activating Nrf2-dependent PANoptosis. Front Pharmacol 2025; 16:1508047. [PMID: 40110131 PMCID: PMC11919867 DOI: 10.3389/fphar.2025.1508047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Background Cisplatin is a widely used drug for the treatment of solid organ cancer, but its renal toxicity cannot be ignored. Amentoflavone (AME), a natural flavonoid compound, has remarkable pharmacological effects, including anti-inflammatory and antioxidative effects. The effect and mechanism of AME on cisplatin-induced acute kidney injury (CI-AKI) remain unclear. Methods We investigated the effect of AME on CI-AKI using the HK-2 cell line and C57BL/6 mice. Renal function, tissue damage, and molecular markers were assessed to explore the effects of AME on oxidative stress and cell death pathways. Results In vitro, AME significantly suppressed the cytotoxic effects of cisplatin on HK-2 cells. Furthermore, AME significantly inhibited cisplatin-induced ferroptosis and PANoptosis (apoptosis, pyroptosis and necroptosis). In mice with acute kidney injury induced by a single intraperitoneal injection of cisplatin, the daily administration of AME during AKI effectively improved renal function and alleviated renal tubular injury, characterized by the normalization of blood urea nitrogen (BUN) and serum creatinine (SCr) levels; it also inhibited cisplatin-induced renal ferroptosis and PANoptosis. AME is a natural antioxidant that activates the Nrf2 antioxidant pathway both in vivo and in vitro. In Nrf2 knockout mice and knockdown cells, the protective effect of AME against cisplatin-induced nephrotoxicity disappeared. However, after Nrf2 knockout, the effect of AME on ferroptosis completely disappeared, and that on PANoptosis partially disappeared. Conclusion Amentoflavone has a protective effect on cisplatin-induced acute kidney injury via a mechanism related to the Nrf2-dependent antioxidant pathway and the regulation of ferroptosis and PANoptosis.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Jianqiang Hu
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Yanmin Zhang
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Xinxin Ci
- Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China
- Jilin Provincial Key Laboratory of Women's Reproductive Health, Changchun, Jilin, China
| |
Collapse
|
46
|
Zhang DD. Thirty years of NRF2: advances and therapeutic challenges. Nat Rev Drug Discov 2025:10.1038/s41573-025-01145-0. [PMID: 40038406 DOI: 10.1038/s41573-025-01145-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2025] [Indexed: 03/06/2025]
Abstract
Over the last 30 years, NRF2 has evolved from being recognized as a transcription factor primarily involved in redox balance and detoxification to a well-appreciated master regulator of cellular proteostasis, metabolism and iron homeostasis. NRF2 plays a pivotal role in diverse pathologies, including cancer, and metabolic, inflammatory and neurodegenerative disorders. It exhibits a Janus-faced duality, safeguarding cellular integrity in normal cells against environmental insults to prevent disease onset, whereas in certain cancers, constitutively elevated NRF2 levels provide a tumour survival advantage, promoting progression, therapy resistance and metastasis. Advances in understanding the mechanistic regulation of NRF2 and its roles in human pathology have propelled the investigation of NRF2-targeted therapeutic strategies. This Review dissects the mechanistic intricacies of NRF2 signalling, its cross-talk with biological processes and its far-reaching implications for health and disease, highlighting key discoveries that have shaped innovative therapeutic approaches targeting NRF2.
Collapse
Affiliation(s)
- Donna D Zhang
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
- University of Florida Health Cancer Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
47
|
Shi M, Li X, Guo Y, Zhang Y, Xu J, Yan L, Liu R, Wang H, Tang S, Zhao Y, Li Z, Feng Y, Ren D, Liu P. Gaudichaudione H Enhances the Sensitivity of Hepatocellular Carcinoma Cells to Disulfidptosis via Regulating NRF2-SLC7A11 Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411131. [PMID: 39840687 PMCID: PMC11923960 DOI: 10.1002/advs.202411131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/01/2024] [Indexed: 01/23/2025]
Abstract
Gaudichaudione H (GH) is a naturally occurring small molecular compound derived from Garcinia oligantha Merr. (Clusiaceae), but the full pharmacological functions remain unclear. Herein, the potential of GH in disulfidptosis regulation, a novel form of programmed cell death induced by disulfide stress is explored. The omics results indicated that NRF2 signaling could be significantly activated by GH. The potential targets are associated with hepatocarcinogenesis and cell death. Moreover, both glutathione (GSH) metabolism and NADP+-NADPH metabolism are affected by GH, indicating the potential in disulfidptosis regulation. It is also observed that GH enhanced the sensitivity of hepatocellular carcinoma (HCC) cells to disulfidptosis, which is dependent on the activation of NRF2-SLC7A11 pathway. GH significantly increased the levels of NRF2 and promoted the transcription of NRF2 target gene, SLC7A11, through autophagy-mediated non-canonical mechanism. Under the condition of glucose starvation, GH-induced upregulation of SLC7A11 aggravated uptake of cysteine, disturbance of GSH synthesis, depletion of NADPH, and accumulation of disulfide molecules, ultimately leading to the formation of disulfide bonds between different cytoskeleton proteins and disulfidptosis eventually. Collectively, the findings underscore the potential role of GH in promoting cancer cell disulfidptosis, thereby offering a promising avenue for the treatment of drug-resistant HCC in clinical settings.
Collapse
Affiliation(s)
- Mengjiao Shi
- Department of General SurgeryNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Xinyan Li
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Ying Guo
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Yinggang Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Jiayi Xu
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Liangwen Yan
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Rongrong Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Hong Wang
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Shenkang Tang
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Department of OncologyAffiliated Hospital of Shaanxi University of Chinese MedicineXianyang712000China
| | - Yaping Zhao
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Zongfang Li
- Department of General SurgeryNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Yetong Feng
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Core Research LaboratoryThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
| | - Dongmei Ren
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Key Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical SciencesShandong UniversityJinan250012China
| | - Pengfei Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and TherapyNational & Local Joint Engineering Research Center of Biodiagnosis and BiotherapyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic DiseasesThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710004China
- Key Laboratory of Environment and Genes Related To DiseasesXi'an Jiaotong UniversityMinistry of Education of ChinaXi'an710061China
| |
Collapse
|
48
|
Liao X, Tang M, Li J, Guo R, Zhong C, Chen X, Zhang X, Mo H, Que D, Yu W, Song X, Li H, Cai Y, Yang P. Acid-Triggered Cascaded Responsive Supramolecular Peptide Alleviates Myocardial Ischemia‒Reperfusion Injury by Restoring Redox Homeostasis and Protecting Mitochondrial Function. Adv Healthc Mater 2025; 14:e2404319. [PMID: 39831810 DOI: 10.1002/adhm.202404319] [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: 11/04/2024] [Revised: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Redox imbalance, including excessive production of reactive oxygen species (ROS) caused by mitochondrial dysfunction and insufficient endogenous antioxidant capacity, is the primary cause of myocardial ischemia‒reperfusion (I/R) injury. In the exploration of reducing myocardial I/R injury, it is found that protecting myocardial mitochondrial function after reperfusion not only reduces ROS bursts but also inhibits cell apoptosis triggered by the release of cytochrome c. Additionally, nuclear factor erythroid 2-related factor 2 (Nrf2) is considered a potential therapeutic target for treating myocardial I/R injury by enhancing the cellular antioxidant capacity through the induction of endogenous antioxidant enzymes. In this study, a peptide‒drug conjugate OI-FFG-ss-SS31(ISP) is developed by integrating the Nrf2 activator 4-octyl itaconate (OI) and the mitochondria-targeting protective peptide elamipretide (SS31), and its therapeutic potential for myocardial I/R injury is explored. The results showed that ISP could self-assemble into nanofibers in response to the acidic microenvironment and bind to Keap-1 with high affinity, thereby activating Nrf2 and enhancing antioxidant capacity. Simultaneously, the release of SS31 could improve mitochondrial function and reduce ROS, ultimately providing a restoration of redox homeostasis to effectively alleviate myocardial I/R injury. This study presents a promising acid-triggered peptide-drug conjugate for treating myocardial I/R injury.
Collapse
Affiliation(s)
- Xu Liao
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Min Tang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Department of Cardiology of Zhuzhou Central Hospital, NO.116 Changjiang South Road Tianyuan District, Zhuzhou, 412000, P. R. China
| | - Jiejing Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Runze Guo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Chongbin Zhong
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Xiangzhou Chen
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Xuwei Zhang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Hongwei Mo
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Dongdong Que
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Wenjie Yu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Xudong Song
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Hekai Li
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Yanbin Cai
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| | - Pingzhen Yang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, P. R. China
- Heart Center of Zhujiang Hospital, Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, 510280, P. R. China
| |
Collapse
|
49
|
Liu H, Zheng S, Hou G, Dai J, Zhao Y, Yang F, Xiang Z, Zhang W, Wang X, Gong Y, Li L, Zhang N, Hu Y. AKAP1/PKA-mediated GRP75 phosphorylation at mitochondria-associated endoplasmic reticulum membranes protects cancer cells against ferroptosis. Cell Death Differ 2025; 32:488-505. [PMID: 39537840 PMCID: PMC11893801 DOI: 10.1038/s41418-024-01414-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Emerging evidence suggests that signaling pathways can be spatially regulated to ensure rapid and efficient responses to dynamically changing local cues. Ferroptosis is a recently defined form of lipid peroxidation-driven cell death. Although the molecular mechanisms underlying ferroptosis are emerging, spatial aspects of its signaling remain largely unexplored. By analyzing a public database, we found that a mitochondrial chaperone protein, glucose-regulated protein 75 (GRP75), may have a previously undefined role in regulating ferroptosis. This was subsequently validated. Interestingly, under ferroptotic conditions, GRP75 translocated from mitochondria to mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) and the cytosol. Further mechanistic studies revealed a highly spatial regulation of GRP75-mediated antiferroptotic signaling. Under ferroptotic conditions, lipid peroxidation predominantly accumulated at the ER, which activated protein kinase A (PKA) in a cAMP-dependent manner. In particular, a signaling microdomain, the outer mitochondrial membrane protein A-kinase anchor protein 1 (AKAP1)-anchored PKA, phosphorylated GRP75 at S148 in MAMs. This caused GRP75 to be sequestered outside the mitochondria, where it competed with Nrf2 for Keap1 binding through a conserved high-affinity RGD-binding motif, ETGE. Nrf2 was then stabilized and activated, leading to the transcriptional activation of a panel of antiferroptotic genes. Blockade of the PKA/GRP75 axis dramatically increased the responses of cancer cells to ferroptosis both in vivo and in vitro. Our identification a localized signaling cascade involved in protecting cancer cells from ferroptosis broadens our understanding of cellular defense mechanisms against ferroptosis and also provides a new target axis (AKAP1/PKA/GRP75) to improve the responses of cancer cells to ferroptosis.
Collapse
Affiliation(s)
- Hao Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China
| | - Shanliang Zheng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Guixue Hou
- BGI-SHENZHEN, Shenzhen, Guangdong Province, 518083, China
| | - Junren Dai
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Yanan Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Fan Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Zhiyuan Xiang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China
| | - Wenxin Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Yafan Gong
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China
| | - Li Li
- The third affiliated hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150040, China
| | - Ning Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China.
- Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, HIT Zhengzhou Research Institute, Zhengzhou, Henan Province, 450000, China.
| |
Collapse
|
50
|
Yang Y, Wang B, Jiang Y, Fu W. Tanshinone IIA mitigates postoperative cognitive dysfunction in aged rats by inhibiting hippocampal inflammation and ferroptosis: Role of Nrf2/SLC7A11/GPX4 axis activation. Neurotoxicology 2025; 107:62-73. [PMID: 39965709 DOI: 10.1016/j.neuro.2025.02.003] [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: 11/21/2024] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
OBJECTIVE Postoperative cognitive dysfunction (POCD) is a common and debilitating complication in elderly patients following surgery, leading to increased morbidity and reduced quality of life. This study aims to investigate the neuroprotective effects of Tanshinone IIA, a lipophilic compound derived from Salvia miltiorrhiza, in an aged rat model of POCD, and explore its underlying molecular mechanisms. METHODS POCD model was established by a modified abdominal exploratory laparotomy. Rats were then intraperitoneally administered with Tanshinone IIA (10 mg/kg, 20 mg/kg, or 40 mg/kg) for 30 days. Cognitive functions were assessed using the morris water maze, novel object recognition test, and Y-maze test. Synaptic structures in the hippocampal CA1 region were examined by electron microscopy. Inflammatory and ferroptosis pathways were evaluated by measuring inflammatory cytokines (TNF-α, IL-6, IL-1β, IL-4), nitric oxide synthase (iNOS) activity, lipid peroxidation products (malondialdehyde [MDA]; 4-hydroxy-2-nonenal [4-HNE]), Fe2 + levels, and antioxidant enzymes (superoxide dismutase [SOD], glutathione [GSH]) using ELISA and commercial kits. mRNA and proteins levels were quantified by real-time quantitative polymerase chain reaction and western blot analysis. RESULTS Tanshinone IIA significantly ameliorated cognitive deficits in aged POCD rats according to behavioral tests. It also restored synaptic ultrastructure in the hippocampal CA1 region and upregulated the expressions of synaptic proteins, including synapsin-1 and PSD-95. In addition, Tanshinone IIA effectively suppressed the hippocampal inflammatory pathway, as evidenced by the decreased levels of pro-inflammatory cytokines (TNF-α, IL-6, IL-1β), an increased level of the anti-inflammatory cytokine IL-4, and the upregulation of the iNOS/NO pathway in the hippocampus. Furthermore, Tanshinone IIA mitigated ferroptosis by reducing MDA and 4-HNE contents, lowering Fe2+ level, and enhancing SOD activity and GSH level. Notably, Tanshinone IIA activated the Nrf2/SLC7A11/GPX4 axis in the hippocampus of aged POCD rats. CONCLUSION These findings suggest that Tanshinone IIA exerts neuroprotective effects in an aged rat model of POCD by attenuating hippocampal inflammation and ferroptosis, primarily through the activation of the Nrf2/SLC7A11/GPX4 axis.
Collapse
Affiliation(s)
- Yan Yang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Bo Wang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Yichen Jiang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China
| | - Wan Fu
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|