1
|
He Y, Zhao G, Ouyang X, Wang S, Chen Y, Li C, He Y, Gao J, Han S, Zhao J, Wang J, Wang C. Creatine-mediated ferroptosis inhibition is involved in the intestinal radioprotection of daytime-restricted feeding. Gut Microbes 2025; 17:2489072. [PMID: 40205678 PMCID: PMC11988229 DOI: 10.1080/19490976.2025.2489072] [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: 10/26/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Ionizing radiation-induced intestinal injury (IRIII) is a catastrophic disease lack of sufficient medical countermeasures currently. Regulation of the gut microbiota through dietary adjustments is a potential strategy to mitigate IRIII. Time-restricted feeding (TRF) is an emerging behavioral nutrition intervention with pleiotropic health benefits. Whether this dietary pattern influences the pathogenesis of IRIII remains vague. We evaluated the impact of TRF on intestinal radiosensitivity in this study and discovered that only daytime TRF (DTRF), not nighttime TRF, could ameliorate intestinal damage in mice that received a high dose of IR. Faecal metagenomic and metabolomic studies revealed that the intestinal creatine level was increased by approximate 9 times by DTRF, to which the Bifidobacterium pseudolongum enrichment contribute. Further investigations showed that creatine could activate the energy sensor AMP-activated protein kinase in irradiated enterocytes and induce phosphorylation of acetyl-CoA carboxylase, resulting in reduced production of polyunsaturated fatty acids and reduced ferroptosis after IR. The administration of creatine mitigated IRIII and reduced bacteremia and proinflammatory responses. Blockade of creatine import compromised the ferroptosis inhibition and mitigation of DTRF on IRIII. Our study demonstrates a radioprotective dietary mode that can reshape the gut microbiota and increase intestinal creatine, which can suppress IR-induced ferroptosis, thereby providing effective countermeasures for IRIII prevention.
Collapse
Affiliation(s)
- Yingjuan He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Gaomei Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Xue Ouyang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Shaobo Wang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yin Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Chenwenya Li
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Yongwu He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jining Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Songling Han
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Miao M, Chen Y, Wang X, Li S, Hu R. The critical role of ferroptosis in virus-associated hematologic malignancies and its potential value in antiviral-antitumor therapy. Virulence 2025; 16:2497908. [PMID: 40302035 PMCID: PMC12045570 DOI: 10.1080/21505594.2025.2497908] [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: 12/03/2024] [Revised: 03/06/2025] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
Epstein-Barr Virus (EBV), Kaposi's sarcoma-associated herpesvirus (KSHV), and human T-cell leukemia virus type 1 (HTLV-1) are key infectious agents linked to the development of various hematological malignancies, including Hodgkin's lymphoma, non-Hodgkin's lymphoma, and adult T-cell leukemia/lymphoma. This review highlights the critical knowledge gaps in understanding the role of ferroptosis, a novel form of cell death, in virus-related tumors. We focus on how ferroptosis influences the host cell response to these viral infections, revealing groundbreaking mechanisms by which the three viruses differentially regulate core pathways of ferroptosis, such as iron homeostasis, lipid peroxidation, and antioxidant systems, thereby promoting malignant transformation of host cells. Additionally, we explore the potential of antiviral drugs and ferroptosis modulators in the treatment of virus-associated hematological malignancies.
Collapse
Affiliation(s)
- Miao Miao
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuelei Chen
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Xuehan Wang
- Shenyang Shenhua Institute Test Technology, Shenyang, Liaoning, China
| | - Shengyang Li
- Publishing Department, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Rong Hu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
3
|
Xiao Y, He M, Zhang X, Yang M, Yuan Z, Yao S, Qin Y. Research progress on the mechanism of tumor cell ferroptosis regulation by epigenetics. Epigenetics 2025; 20:2500949. [PMID: 40327848 DOI: 10.1080/15592294.2025.2500949] [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: 02/14/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer remains a significant barrier to human longevity and a leading cause of mortality worldwide. Despite advancements in cancer therapies, challenges such as cellular toxicity and drug resistance to chemotherapy persist. Regulated cell death (RCD), once regarded as a passive process, is now recognized as a programmed mechanism with distinct biochemical and morphological characteristics, thereby presenting new therapeutic opportunities. Ferroptosis, a novel form of RCD characterized by iron-dependent lipid peroxidation and unique mitochondrial damage, differs from apoptosis, autophagy, and necroptosis. It is driven by reactive oxygen species (ROS)-induced lipid peroxidation and is implicated in tumorigenesis, anti-tumor immunity, and resistance, particularly in tumors undergoing epithelial-mesenchymal transition. Moreover, ferroptosis is associated with ischemic organ damage, degenerative diseases, and aging, regulated by various cellular metabolic processes, including redox balance, iron metabolism, and amino acid, lipid, and glucose metabolism. This review focuses on the role of epigenetic factors in tumor ferroptosis, exploring their mechanisms and potential applications in cancer therapy. It synthesizes current knowledge to provide a comprehensive understanding of epigenetic regulation in tumor cell ferroptosis, offering insights for future research and clinical applications.
Collapse
Affiliation(s)
- Yuyang Xiao
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mengyang He
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xupeng Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Meng Yang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhangchi Yuan
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shanhu Yao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Medical Information Research, Central South University, Changsha, Hunan, China
| | - Yuexiang Qin
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Lyu G, Liao H, Li R. Ferroptosis and renal fibrosis: mechanistic insights and emerging therapeutic targets. Ren Fail 2025; 47:2498629. [PMID: 40329437 DOI: 10.1080/0886022x.2025.2498629] [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: 01/04/2025] [Revised: 04/01/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025] Open
Abstract
Ferroptosis is a regulated, iron-dependent form of cell death driven by lipid peroxidation and distinct from apoptosis, necroptosis, and pyroptosis. Recent studies implicate ferroptosis as a central contributor to the pathogenesis of renal fibrosis, a hallmark of chronic kidney disease associated with high morbidity and progression to end-stage renal failure. This review synthesizes current evidence linking ferroptotic signaling to fibrotic remodeling in the kidney, focusing on iron metabolism dysregulation, glutathione peroxidase 4 (GPX4) inactivation, lipid peroxide accumulation, and ferroptosis-regulatory pathways such as FSP1-CoQ10-NAD(P)H and GCH1-BH4. We detail how ferroptosis in tubular epithelial cells modulates pro-fibrotic cytokine release, macrophage recruitment, and TGF-β1-driven extracellular matrix deposition. Moreover, we explore ferroptosis as a therapeutic vulnerability in renal fibrosis, highlighting promising agents including iron chelators, GPX4 activators, anti-lipid peroxidants, and exosome-based gene delivery systems. By consolidating emerging preclinical data, this review provides a comprehensive mechanistic framework and identifies translational opportunities for targeting ferroptosis in fibrotic kidney disease.
Collapse
Affiliation(s)
- Guangna Lyu
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
- The Second People's Hospital of Shanxi Province, Taiyuan, China
| | - Hui Liao
- The Drug Clinical Trial Institution of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | - Rongshan Li
- The Nephrology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
5
|
Ren X, Zhao L, Hao Y, Huang X, Lv G, Zhou X. Copper-instigated modulatory cell mortality mechanisms and progress in kidney diseases. Ren Fail 2025; 47:2431142. [PMID: 39805816 PMCID: PMC11734396 DOI: 10.1080/0886022x.2024.2431142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/23/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025] Open
Abstract
Copper is a vital cofactor in various enzymes, plays a pivotal role in maintaining cell homeostasis. When copper metabolism is disordered and mitochondrial dysfunction is impaired, programmed cell death such as apoptosis, paraptosis, pyroptosis, ferroptosis, cuproptosis, autophagy and necroptosis can be induced. In this review, we focus on the metabolic mechanisms of copper. In addition, we discuss the mechanism by which copper induces various programmed cell deaths. Finally, this review examines copper's involvement in prevalent kidney diseases such as acute kidney injury and chronic kidney disease. The findings indicate that the use of copper chelators or plant extracts can mitigate kidney damage by reducing copper accumulation, offering novel insights into the pathogenesis and treatment strategies for kidney diseases.
Collapse
Affiliation(s)
- Xiya Ren
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Limei Zhao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yajie Hao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiu Huang
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guangna Lv
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoshuang Zhou
- Department of Nephrology, Shanxi Provincial People’s Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
6
|
Zheng Q, Wang D, Lin R, Xu W. Pyroptosis, ferroptosis, and autophagy in spinal cord injury: regulatory mechanisms and therapeutic targets. Neural Regen Res 2025; 20:2787-2806. [PMID: 39101602 PMCID: PMC11826477 DOI: 10.4103/nrr.nrr-d-24-00112] [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: 01/28/2024] [Revised: 04/24/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Regulated cell death is a form of cell death that is actively controlled by biomolecules. Several studies have shown that regulated cell death plays a key role after spinal cord injury. Pyroptosis and ferroptosis are newly discovered types of regulated cell deaths that have been shown to exacerbate inflammation and lead to cell death in damaged spinal cords. Autophagy, a complex form of cell death that is interconnected with various regulated cell death mechanisms, has garnered significant attention in the study of spinal cord injury. This injury triggers not only cell death but also cellular survival responses. Multiple signaling pathways play pivotal roles in influencing the processes of both deterioration and repair in spinal cord injury by regulating pyroptosis, ferroptosis, and autophagy. Therefore, this review aims to comprehensively examine the mechanisms underlying regulated cell deaths, the signaling pathways that modulate these mechanisms, and the potential therapeutic targets for spinal cord injury. Our analysis suggests that targeting the common regulatory signaling pathways of different regulated cell deaths could be a promising strategy to promote cell survival and enhance the repair of spinal cord injury. Moreover, a holistic approach that incorporates multiple regulated cell deaths and their regulatory pathways presents a promising multi-target therapeutic strategy for the management of spinal cord injury.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Weihong Xu
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
7
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [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: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
8
|
Peng W, Liang J, Qian X, Li M, Nie M, Chen B. IGF2BP1/AIFM2 axis regulates ferroptosis and glycolysis to drive hepatocellular carcinoma progression. Cell Signal 2025; 130:111660. [PMID: 39971223 DOI: 10.1016/j.cellsig.2025.111660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is aggressive liver tumor that is the third leading cause of cancer death. Ferroptosis and glycolysis play key roles in HCC progression. Apoptosis-inducing factor mitochondria-associated 2 (AIFM2) in involved in regulating ferroptosis and glycolysis in cancers, but its role in HCC remains unclear. This research explored the function of AIFM2 in HCC. METHODS AIFM2 expression in HCC tissues was evaluated using the UALCAN and GEPIA databases, as well as RT-qPCR. Kaplan-Meier survival analysis analyzed the correlation between AIFM2 and the prognosis of HCC patients. EdU and transwell assays were utilized to examine HCC cell proliferation, migration, and invasion. Ferroptosis markers were analyzed by measuring iron levels, ROS production (DCFH-DA assay), and oxidative stress indicators (SOD, MDA, and GSH). Glycolytic activity was assessed through glucose uptake, lactate production, and ATP levels. m6A modification on AIFM2 mRNA was confirmed by MeRIP assay, and mRNA stability was evaluated with Actinomycin D treatment. Tumor growth and metastasis were studied in xenograft and lung metastasis models. RESULTS UALCAN analysis showed that AIFM2 was significantly upregulated in HCC tissues, which correlated with poor survival rates of HCC patients. IGF2BP1 was also highly expressed in HCC tissues and positively correlated with AIFM2 levels in HCC tissues. Functionally, AIFM2 knockdown suppressed glycolysis and enhanced ferroptosis, while its overexpression had opposite effects. IGF2BP1 was found to stabilize AIFM2 mRNA via m6A modification, promoting AIFM2 expression. IGF2BP1 knockdown reduced glycolysis, proliferation, and invasion while promoting ferroptosis, while AIFM2 overexpression could reverse this effect. In vivo, IGF2BP1 or AIFM2 silencing significantly suppressed tumor growth and metastasis. CONCLUSION IGF2BP1 stabilized AIFM2 mRNA to regulate ferroptosis and glycolysis and promoted HCC progression.
Collapse
Affiliation(s)
- Wei Peng
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Jie Liang
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Xuanlv Qian
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Mingwang Li
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Ming Nie
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China
| | - Bin Chen
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital Bijie Hospital, Bijie, Guizhou 551700, China.
| |
Collapse
|
9
|
Gong L, Wu L, Zhao S, Xiao S, Chu X, Zhang Y, Li F, Li S, Yang H, Jiang P. Epigenetic regulation of ferroptosis in gastrointestinal cancers (Review). Int J Mol Med 2025; 55:93. [PMID: 40242977 PMCID: PMC12045471 DOI: 10.3892/ijmm.2025.5534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Ferroptosis is a type of iron‑dependent cell death characterized by excessive lipid peroxidation and may serve as a potential therapeutic target in cancer treatment. While the mechanisms governing ferroptosis continue to be explored and elucidated, an increasing body of research highlights the significant impact of epigenetic modifications on the sensitivity of cancer cells to ferroptosis. Epigenetic processes, such as DNA methylation, histone modifications and non‑coding RNAs, have been identified as key regulators that modulate the expression of ferroptosis‑related genes. These alterations can either enhance or inhibit the sensitivity of gastrointestinal cancer (GIC) cells to ferroptosis, thereby affecting the fate of GICs. Drugs that target epigenetic markers for advanced‑stage cancer have shown promising results in enhancing ferroptosis and inhibiting tumor growth. This review explores the intricate relationship between epigenetic regulation and ferroptosis in GICs. Additionally, the potential of leveraging epigenetic modifications to trigger ferroptosis in GICs is investigated. This review highlights the importance of further research to elucidate the specific mechanisms underlying epigenetic control of ferroptosis and to advance the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Linqiang Gong
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Linlin Wu
- Oncology Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shiyuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| | - Shuai Xiao
- Department of Intensive Care Medicine, Tengzhou Central People's Hospital, Jining Medical University, Tengzhou, Shandong 277500, P.R. China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
| | - Yazhou Zhang
- Department of Foot and Ankle Surgery, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Fengfeng Li
- Neurosurgery Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shuhui Li
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Hui Yang
- Department of Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| |
Collapse
|
10
|
Yang J, Wang Y, Liu F, Zhang Y, Han F. Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review). Int J Mol Med 2025; 55:97. [PMID: 40314096 PMCID: PMC12045474 DOI: 10.3892/ijmm.2025.5538] [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: 01/27/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025] Open
Abstract
Ferroptosis is a unique mode of cell death driven by iron‑dependent phospholipid peroxidation, and its mechanism primarily involves disturbances in iron metabolism, imbalances in the lipid antioxidant system and accumulation of lipid peroxides. Protein processing, modification and folding in the endoplasmic reticulum (ER) are closely related regulatory processes that determine cell function, fate and survival. The uncontrolled proliferative capacity of malignant cells generates an unfavorable microenvironment characterized by high metabolic demand, hypoxia, nutrient deprivation and acidosis, which promotes the accumulation of misfolded or unfolded proteins in the ER, leading to ER stress (ERS). Ferroptosis and ERS share common pathways in several diseases, and the two interact to affect cell survival and death. Additionally, cell death pathways are not linear signaling cascades, and different pathways of cell death may be interrelated at multiple levels. Ferroptosis and ERS in ovarian cancer (OC) have attracted increasing research interest; however, both are discussed separately regarding OC. The present review aims to summarize the associations and potential links between ferroptosis and ERS, aiming to provide research references for the development of therapeutic approaches for the management of OC.
Collapse
Affiliation(s)
- Jiaqi Yang
- Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yu Wang
- Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Fangyuan Liu
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yizhong Zhang
- Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Fengjuan Han
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
11
|
Wang L, Yu H, Wang D, Yin G, Chen S, Zhang X, Yu W, Meng D, Liu H, Jiang W, Zhang F. Diosgenin alleviates lipid accumulation in NAFLD through the pathways of ferroptosis defensive and executive system. J Nutr Biochem 2025; 140:109886. [PMID: 40023201 DOI: 10.1016/j.jnutbio.2025.109886] [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: 08/26/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
The most prevalent liver condition globally is non-alcoholic fatty liver disease (NAFLD), for which no approved therapies currently exist. Diosgenin, an important component in plants from the Leguminosae, Dioscoreaceae, and Solanaceae families, has demonstrated considerable anti-inflammatory and antioxidant effects. Nonetheless, the specific mechanism by which it may act in managing NAFLD remains unclear. Our research aims to explore the effects and molecular mechanisms of DG on NAFLD by utilizing both in vivo and in vitro experimental approaches. To investigate the effect of DG on hepatic steatosis, we used Sprague-Dawley rats induced by a high-fat diet (HFD) and HepG2 cells exposed to free fatty acids. Oil red O staining and hematoxylin-eosin (H&E) staining were used to explore lipid accumulation and hepatic degeneration. ROS staining, SOD, MDA, and Fe2+kits were used to detect the indexes related to oxidative stress in ferroptosis in hepatic tissues and cells. IFSP1 and pcDNA3.1-ACSL4 plasmid were used to knock down Ferroptosis suppressor protein1 (FSP1) and promote the expression of acyl-CoA synthetase long-chain family member 4 (ACSL4) in HepG2 cells. DG improved lipid metabolism disorders and liver damage induced by a high-fat diet in rats with NAFLD. Furthermore, the administration of DG notably decreased oxidative stress levels and liver Fe2+ concentrations in rats. Additionally, in vitro experiments demonstrated that DG treatment markedly attenuated ferroptosis and ROS accumulation in HepG2 cells induced by FFAs. Moreover, overexpression of hepatic ACSL4 expression by pcDNA3.1-ACSL4 plasmid promoted the regulatory effects of DG on LPCAT3 and ALOX15. Our research shows that DG can alleviate NAFLD by regulating the FSP1/COQ10 pathway of the ferroptosis defense system and the ACSL4/LPCAT3/ALOX15 pathway of the ferroptosis execution system. Therefore, DG may serve as a novel inhibitor of ferroptosis for the treatment of NAFLD.
Collapse
Affiliation(s)
- Linya Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongzhuan Yu
- Weifang Traditional Chinese Medicine Hospital, Shandong, China
| | - Dongxian Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoliang Yin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Suwen Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfei Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Decheng Meng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongshuai Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenying Jiang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fengxia Zhang
- Department of Neurology, the first Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong, China.
| |
Collapse
|
12
|
Zhang H, Tian L, Wang P, Li L, Wang K, Li Y, Zhang Y, Feng L, Yao S, Guan H, Ren W. Ferrostatin-1 mitigates acute lung injury by reducing ferroptosis levels in gas explosions. Tissue Cell 2025; 94:102773. [PMID: 39954560 DOI: 10.1016/j.tice.2025.102773] [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: 11/26/2024] [Revised: 01/15/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Gas explosion injuries are a severe form of trauma with high incidence and mortality rates, both in daily life and industrial settings. Acute lung injury (ALI) is one of the most serious complications of gas explosion injuries and is a leading cause of mortality in such cases. However, the mechanisms underlying gas explosion-induced ALI have not been fully elucidated, and the treatment process consumes a significant amount of medical resources. Therefore, it is crucial to conduct research on the injury mechanisms of gas explosion injuries, especially the mechanisms of gas explosion-induced ALI, which can effectively improve the treatment rate of this condition. In this study, we analyzed the relationship between a novel form of cell death, ferroptosis, and gas explosion-induced ALI, and explored its specific mechanisms. METHODS We established ALI rat model by Shock tube biological injury system, and detected lung injury-related indexes as well as ferroptosis related indexes, such as glutathione peroxidase 4(GPX4), 4-hydroxynonenal(4HNE), Malondialdehyde(MDA), Fe2 + . We also investigated the therapeutic effects of the ferroptosis inhibitor ferrostatin-1 (Fer-1) in ALI induced by gas explosion, as well as its specific mechanisms of action. RESULTS A rat ALI model by gas explosion was successfully established. After the gas explosion treatment, we observed that the systemic inflammatory reaction of rats was increased, and lung function, liver function, kidney function and cardiac function were damaged to different degrees. The inflammatory infiltration in the lung tissue was more severe, and the degree of lung injury and pulmonary edema increased. The ferroptosis markers GPX4 was decreased, while the levels of 4HNE, MDA and Fe2 + were increased. Treatment with Fer-1 significantly ameliorated gas explosion ALI damage and down-regulated the expression level of ferroptosis. CONCLUSIONS Gas explosion-induced ALI in rats is characterized by enhanced inflammatory responses and reduced antioxidant capacity in lung tissues. The specific mechanism of injury involves ferroptosis. Fer-1 has been shown to mitigate the severity of ALI caused by gas explosion by suppressing ferroptosis expression levels in lung tissues via the Nrf2/GPX4 axis.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; Department of Orthopedics of the General Hospital of Western Theater Command, Chengdu 610083, China
| | - Linqiang Tian
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Peng Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Long Li
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China
| | - Kunxi Wang
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Yanyan Li
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Yue Zhang
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Lili Feng
- Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; School of Public Health, Xinxiang Medical University, Xinxiang 453003, China.
| | - Hao Guan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Wenjie Ren
- Institutes of Health Central Plain, Xinxiang Medical University, Jinhui Road #601, Xinxiang, Henan 453003, China; Institute of Trauma Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China; Clinical Medical Center of Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang 453003, China.
| |
Collapse
|
13
|
Zhang Z, Yang J, Zhou Q, Zhong S, Liu J, Zhang X, Chang X, Wang H. The cGAS-STING-mediated ROS and ferroptosis are involved in manganese neurotoxicity. J Environ Sci (China) 2025; 152:71-86. [PMID: 39617588 DOI: 10.1016/j.jes.2024.05.003] [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: 01/31/2024] [Revised: 04/12/2024] [Accepted: 05/01/2024] [Indexed: 12/12/2024]
Abstract
Manganese (Mn) has been characterized as an environmental pollutant. Excessive releases of Mn due to human activities have increased Mn levels in the environment over the years, posing a threat to human health and the environment. Long-term exposure to high concentrations of Mn can induce neurotoxicity. Therefore, toxicological studies on Mn are of paramount importance. Mn induces oxidative stress through affecting the level of reactive oxygen species (ROS), and the overabundance of ROS further triggers ferroptosis. Additionally, Mn2+ was found to be a novel activator of the cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway in the innate immune system. Thus, we speculate that Mn exposure may promote ROS production by activating the cGAS-STING pathway, which further induces oxidative stress and ferroptosis, and ultimately triggers Mn neurotoxicity. This review discusses the mechanism between Mn-induced oxidative stress and ferroptosis via activation of the cGAS-STING pathway, which may offer a prospective direction for future in-depth studies on the mechanism of Mn neurotoxicity.
Collapse
Affiliation(s)
- Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
14
|
Catapano A, Cimmino F, Petrella L, Pizzella A, D'Angelo M, Ambrosio K, Marino F, Sabbatini A, Petrelli M, Paolini B, Lucchin L, Cavaliere G, Cristino L, Crispino M, Trinchese G, Mollica MP. Iron metabolism and ferroptosis in health and diseases: The crucial role of mitochondria in metabolically active tissues. J Nutr Biochem 2025; 140:109888. [PMID: 40057002 DOI: 10.1016/j.jnutbio.2025.109888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/30/2025]
Abstract
Iron is essential in various physiological processes, but its accumulation leads to oxidative stress and cell damage, thus iron homeostasis has to be tightly regulated. Ferroptosis is an iron-dependent non-apoptotic regulated cell death characterized by iron overload and reactive oxygen species accumulation. Mitochondria are organelles playing a crucial role in iron metabolism and involved in ferroptosis. MitoNEET, a protein of mitochondrial outer membrane, is a key element in this process. Ferroptosis, altering iron levels in several metabolically active organs, is linked to several non-communicable diseases. For example, iron overload in the liver leads to hepatic fibrosis and cirrhosis, accelerating non-alcholic fatty liver diseases progression, in the muscle cells contributes to oxidative damage leading to sarcopenia, and in the brain is associated to neurodegeneration. The aim of this review is to investigate the intricate balance of iron regulation focusing on the role of mitochondria and oxidative stress, and analyzing the ferroptosis implications in health and disease.
Collapse
Affiliation(s)
- Angela Catapano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy; Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Lidia Petrella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Katia Ambrosio
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Francesca Marino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Annarita Sabbatini
- Dietetic and Clinical Nutrition Unit, IEO European Institute of Oncology IRCSS, Milan, Italy
| | - Massimiliano Petrelli
- Department of Clinical and Molecular Sciences, Clinic of Endocrinology and Metabolic Diseases, Università Politecnica delle Marche, Ancona, Italy
| | - Barbara Paolini
- Department of Innovation, experimentation and clinical research, Unit of dietetics and clinical nutrition, S. Maria Alle Scotte Hospital, University of Siena, Siena, Italy
| | - Lucio Lucchin
- Dietetics and Clinical Nutrition, Bolzano Health District, Bolzano, Italy
| | - Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | | | | |
Collapse
|
15
|
Gu D, Liang X, Shao Z, He H, Zhu L, Qiu S, Liu Z, Wang S, Chen H, Ci S, Wu H, Wang Y. A cascaded amplification carrier-free nanoplatform for synergistic photothermal/ferroptosis therapy via dual antioxidant pathway disruption in cervical cancer. J Mater Chem B 2025. [PMID: 40337781 DOI: 10.1039/d5tb00627a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Cellular defense mechanisms against ferroptosis are primarily mediated by antiferroptotic regulators, particularly glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1). Notably, singlet oxygen (1O2) generated through photoactivation of organic small-molecule photosensitizers (PSs) has been demonstrated to deplete both glutathione (GSH) and nicotinamide adenine dinucleotide phosphate (NADPH). This dual depletion mechanism effectively disrupts the GSH/GPX4 redox axis and the NADPH/FSP1/ubiquinone (CoQ) antioxidant system, thereby potentiating ferroptosis. In this study, we engineered a tumor-targeting amphiphilic iridium-based photosensitizer nanoplatform (Ir-TCF3P-FA NPs) for synergistic photothermal-ferroptosis therapy. Specifically, GSH depletion and NADPH oxidation by 1O2 produced via Ir-TCF3P-FA NPs at 450 nm can suppress the expression of GPX4 and FSP1, amplifying ferroptosis. Additionally, TCF3P exhibited high photothermal conversion efficiency at 808 nm, which not only can enhance photothermal therapy (PTT) efficacy but also facilitated 1O2 generation. The Ir-TCF3P-FA NPs enable effective tumor-targeted delivery and fluorescence/photoacoustic imaging for in vivo distribution tracking. In vivo studies revealed that dual-laser irradiation of Ir-TCF3P-FA NPs provided potent therapeutic efficacy, significantly inhibiting human cervical cancer progression in murine models. This cascaded amplification carrier-free nanoplatform holds promise for clinical multimodal treatment of cervical cancer.
Collapse
Affiliation(s)
- Dihai Gu
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China.
| | - Xiao Liang
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Zitong Shao
- The First Clinical Medical College, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Haonan He
- The First Clinical Medical College, Nanjing Medical University, Nanjing 210029, P. R. China
| | - Lin Zhu
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Shali Qiu
- Department of Pathology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Zhen Liu
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Senlin Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China.
| | - Haijiao Chen
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Shusheng Ci
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, P. R. China
| | - Hongshuai Wu
- Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Oncology, Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, P. R. China.
| | - Yihong Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210089, P. R. China.
| |
Collapse
|
16
|
Cañeque T, Baron L, Müller S, Carmona A, Colombeau L, Versini A, Solier S, Gaillet C, Sindikubwabo F, Sampaio JL, Sabatier M, Mishima E, Picard-Bernes A, Syx L, Servant N, Lombard B, Loew D, Zheng J, Proneth B, Thoidingjam LK, Grimaud L, Fraser CS, Szylo KJ, Der Kazarian E, Bonnet C, Charafe-Jauffret E, Ginestier C, Santofimia-Castaño P, Estaras M, Dusetti N, Iovanna JL, Cunha AS, Pittau G, Hammel P, Tzanis D, Bonvalot S, Watson S, Gandon V, Upadhyay A, Pratt DA, Freitas FP, Friedmann Angeli JP, Stockwell BR, Conrad M, Ubellacker JM, Rodriguez R. Activation of lysosomal iron triggers ferroptosis in cancer. Nature 2025:10.1038/s41586-025-08974-4. [PMID: 40335696 DOI: 10.1038/s41586-025-08974-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/03/2025] [Indexed: 05/09/2025]
Abstract
Iron catalyses the oxidation of lipids in biological membranes and promotes a form of cell death called ferroptosis1. Defining where this chemistry occurs in the cell can inform the design of drugs capable of inducing or inhibiting ferroptosis in various disease-relevant settings. Genetic approaches have revealed suppressors of ferroptosis2-4; by contrast, small molecules can provide spatiotemporal control of the chemistry at work5. Here we show that the ferroptosis inhibitor liproxstatin-1 exerts cytoprotective effects by inactivating iron in lysosomes. We also show that the ferroptosis inducer RSL3 initiates membrane lipid oxidation in lysosomes. We designed a small-molecule activator of lysosomal iron-fentomycin-1-to induce the oxidative degradation of phospholipids and ultimately ferroptosis. Fentomycin-1 is able to kill iron-rich CD44high primary sarcoma and pancreatic ductal adenocarcinoma cells, which can promote metastasis and fuel drug tolerance. In such cells, iron regulates cell adaptation6,7 while conferring vulnerability to ferroptosis8,9. Sarcoma cells exposed to sublethal doses of fentomycin-1 acquire a ferroptosis-resistant cell state characterized by the downregulation of mesenchymal markers and the activation of a membrane-damage response. This phospholipid degrader can eradicate drug-tolerant persister cancer cells in vitro and reduces intranodal tumour growth in a mouse model of breast cancer metastasis. Together, these results show that control of iron reactivity confers therapeutic benefits, establish lysosomal iron as a druggable target and highlight the value of targeting cell states10.
Collapse
Affiliation(s)
- Tatiana Cañeque
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Leeroy Baron
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Sebastian Müller
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Alanis Carmona
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Ludovic Colombeau
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Antoine Versini
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Stéphanie Solier
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
- Department of Genetics, Institut Curie, Paris, France
- Paris Saclay University, UVSQ, Montigny-le-Bretonneux, France
| | - Christine Gaillet
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | | | - Julio L Sampaio
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Marie Sabatier
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Laurène Syx
- Institut Curie, INSERM, Mines Paris Tech, PSL Research University, Paris, France
| | - Nicolas Servant
- Institut Curie, INSERM, Mines Paris Tech, PSL Research University, Paris, France
| | | | - Damarys Loew
- Institut Curie, PSL Research University, Paris, France
| | - Jiashuo Zheng
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Laurence Grimaud
- Ecole Normale Supérieure, CNRS, PSL Research University, Paris, France
| | - Cameron S Fraser
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Krystina J Szylo
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Emma Der Kazarian
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Caroline Bonnet
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | | | - Christophe Ginestier
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Patricia Santofimia-Castaño
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Matias Estaras
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Nelson Dusetti
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Juan Lucio Iovanna
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Antonio Sa Cunha
- Department of Digestive and Medical Oncology, Paris Saclay University and Paul-Brousse Hospital (APHP Sud), Villejuif, France
| | - Gabriella Pittau
- Department of Digestive and Medical Oncology, Paris Saclay University and Paul-Brousse Hospital (APHP Sud), Villejuif, France
| | - Pascal Hammel
- Department of Digestive and Medical Oncology, Paris Saclay University and Paul-Brousse Hospital (APHP Sud), Villejuif, France
| | - Dimitri Tzanis
- Department of Surgical Oncology, Institut Curie, Paris, France
| | - Sylvie Bonvalot
- Department of Surgical Oncology, Institut Curie, Paris, France
| | - Sarah Watson
- Department of Medical Oncology, Institut Curie, INSERM, PSL Research University, Paris, France
| | - Vincent Gandon
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, CNRS, Paris Saclay University, Orsay, France
| | - Aditya Upadhyay
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Florêncio Porto Freitas
- Rudolf-Virchow-Zentrum, Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - José Pedro Friedmann Angeli
- Rudolf-Virchow-Zentrum, Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, USA
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, TUM Natural School of Sciences, Technical University of Munich, Garching, Germany
| | - Jessalyn M Ubellacker
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Raphaël Rodriguez
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France.
| |
Collapse
|
17
|
Xu M, Xu B. Protein lipidation in the tumor microenvironment: enzymology, signaling pathways, and therapeutics. Mol Cancer 2025; 24:138. [PMID: 40335986 DOI: 10.1186/s12943-025-02309-7] [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: 11/27/2024] [Accepted: 03/18/2025] [Indexed: 05/09/2025] Open
Abstract
Protein lipidation is a pivotal post-translational modification that increases protein hydrophobicity and influences their function, localization, and interaction network. Emerging evidence has shown significant roles of lipidation in the tumor microenvironment (TME). However, a comprehensive review of this topic is lacking. In this review, we present an integrated and in-depth literature review of protein lipidation in the context of the TME. Specifically, we focus on three major lipidation modifications: S-prenylation, S-palmitoylation, and N-myristoylation. We emphasize how these modifications affect oncogenic signaling pathways and the complex interplay between tumor cells and the surrounding stromal and immune cells. Furthermore, we explore the therapeutic potential of targeting lipidation mechanisms in cancer treatment and discuss prospects for developing novel anticancer strategies that disrupt lipidation-dependent signaling pathways. By bridging protein lipidation with the dynamics of the TME, our review provides novel insights into the complex relationship between them that drives tumor initiation and progression.
Collapse
Affiliation(s)
- Mengke Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China.
| |
Collapse
|
18
|
Liang W, Guo H, Li L, Tan W, Liu J, Hu X, Wang Y, Zhou S. Ferroptosis: a new target for depression prevention and treatment. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02912-4. [PMID: 40317298 DOI: 10.1007/s00702-025-02912-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/06/2025] [Indexed: 05/07/2025]
Abstract
Depression, a significant mental health issue, is one of the diseases with the highest disability rates worldwide. The exact etiology of depression remains undetermined, complicating the development of treatment strategies targeting specific mechanisms, and there is currently no effective cure. In this context, ferroptosis may represent a breakthrough in the understanding of depression. Ferroptosis is primarily associated with iron accumulation and lipid peroxidation, and recent studies have revealed its potential association with depression. Clinical evidence suggests that ferroptosis may influence the development and function of the hippocampus through interactions with neuroinflammation. Activated microglia, astrocytes, and neurons are involved in ferroptosis. This review summarizes recent findings on how ferroptosis contributes to depression, including glutathione peroxidase 4 (GPX4), nuclear factor-erythroid 2-related factor 2 (Nrf2), phase separation, and neuroinflammatory pathways, allowing the proposal of some new hypotheses. We hope that exploring the role of ferroptosis in the mechanism of depression will offer a new perspective on the complex biological basis of depression and provide theoretical support for the development of new therapeutic methods.
Collapse
Affiliation(s)
- Wenxuan Liang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China
- Basic Medical College, Guilin Medical College, Guilin, 541199, China
| | - Haowei Guo
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China
- Basic Medical College, Guilin Medical College, Guilin, 541199, China
| | - Luyao Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China
- Basic Medical College, Guilin Medical College, Guilin, 541199, China
| | - Wupeng Tan
- Department of Gynaecology, Maternal and Child Health Hospital of Hengyang, Hengyang, 421001, China
| | - Jianfeng Liu
- Department of Pediatrics, Second Affiliated Hospital of South China University, Hengyang, 421001, China
| | - Xiaoli Hu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China
- Basic Medical College, Guilin Medical College, Guilin, 541199, China
| | - Yuchu Wang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China.
- Basic Medical College, Guilin Medical College, Guilin, 541199, China.
| | - Shouhong Zhou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, 541199, China.
- Basic Medical College, Guilin Medical College, Guilin, 541199, China.
| |
Collapse
|
19
|
Demuynck R, Saviuk M, Krysko DV. Palmitoylation regulates ferroptosis and anti-tumor immunity. NATURE CANCER 2025:10.1038/s43018-025-00972-9. [PMID: 40316767 DOI: 10.1038/s43018-025-00972-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Affiliation(s)
- Robin Demuynck
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Mariia Saviuk
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy and Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
20
|
Xiao W, Yike W, Gongwen L, Youjia X. Ferroptosis-mediated immune responses in osteoporosis. J Orthop Translat 2025; 52:116-125. [PMID: 40271049 PMCID: PMC12017889 DOI: 10.1016/j.jot.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Osteoporosis is a common systemic metabolic disease, characterized by decreased bone mass and susceptibility to fragility fractures, often associated with aging, menopause, genetics, and immunity. Ferroptosis plays an underestimated yet crucial role in the further impact of immune function changes on osteoporosis. Cell ferroptosis can induce alterations in immune function, subsequently influencing bone metabolism. In this context, this review summarizes several mechanisms of ferroptosis and introduces the latest insights on how ferroptosis regulates immune responses, exploring the interactions between ferroptosis and other mechanisms such as oxidative stress, inflammation, etc. This review elucidates potential treatment strategies for osteoporosis, emphasizing the promising potential of ferroptosis as an emerging target in the treatment of osteoporosis. In conclusion, preparations related to ferroptosis exhibit substantial clinical promise for enhancing bone mass restoration. The translational potential of this article: This review elucidates a nuanced conversation between the immune system and osteoporosis, with ferroptosis serving as the connecting link. These findings underscore the potential of ferroptosis inhibition as a therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Wang Xiao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wang Yike
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liu Gongwen
- Department of Orthopaedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xu Youjia
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
21
|
Peng T, Li M. Research progress of traditional Chinese medicines in regulating acute kidney injury-related ferroptosis: a literature review. Int Urol Nephrol 2025; 57:1601-1608. [PMID: 39680293 PMCID: PMC12003539 DOI: 10.1007/s11255-024-04302-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/20/2024] [Indexed: 12/17/2024]
Abstract
Ferroptosis plays a significant role in the pathological mechanism of acute kidney injury (AKI) for many etiologies. The characteristics of ferroptosis involve many aspects, including iron metabolism, lipid metabolism, and glutathione metabolism. In terms of iron metabolism, ferroptosis involves the accumulation of labile iron; in terms of lipid metabolism, ferroptosis involves the peroxidation of lipids, especially certain phospholipids; in terms of glutathione metabolism, ferroptosis involves the reduction of reduced glutathione (GSH) levels, leading to a decrease in the activity of glutathione peroxidase 4 (GPX4). A lot of traditional Chinese medicines (TCMs) have been reported to have a protective effect against AKI, and many of these TCMs have shown a close association with regulating ferroptosis in ameliorating AKI. While the mechanisms through which these TCMs regulate ferroptosis associated with AKI are intricate, many of their targets are linked to the inhibition of lipid peroxidation or the regulation of iron metabolism. This article discusses some aspects of AKI and ferroptosis, and reviews some research progress on the regulation of AKI-related ferroptosis by TCMs.
Collapse
Affiliation(s)
- Tao Peng
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Mingquan Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
22
|
Yang Y, Shen W, Zhang Z, Dai Y, Zhang Z, Liu T, Yu J, Huang S, Ding Y, You R, Wang Z, Wu Y, Bian T. FSP1 Acts in Parallel with GPX4 to Inhibit Ferroptosis in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2025; 72:551-562. [PMID: 39453438 PMCID: PMC12051924 DOI: 10.1165/rcmb.2023-0467oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 10/25/2024] [Indexed: 10/26/2024] Open
Abstract
GPX4 (glutathione peroxidase 4) has recently been reported to play an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD). FSP1 (ferroptosis suppressor protein-1) is a protein that defends against ferroptosis in parallel with GPX4, but its role in the pathogenesis of COPD remains unexplored, and further research is needed. Normal and COPD lung tissues were obtained from lobectomy and lung transplant specimens, respectively. FSP1-overexpressing mice were established by monthly transfection with adenoassociated virus 9-FSP1 through modified intranasal administration. The expression of FSP1, GPX4, and PTGS2 (prostaglandin-endoperoxide synthase 2) was measured by Western blotting, immunohistochemistry and other methods. The correlation between FSP1 and ferroptosis and the role of FSP1 in COPD were explored by screening the expression of ferroptosis-related genes in a COPD cell model after the inhibition and overexpression of FSP1. We then explored the underlying mechanism of low FSP1 expression in patients with COPD in vitro by methylated RNA immunoprecipitation quantitative qPCR. We found that cigarette smoke exposure can lead to an increase in lipid peroxide production and ultimately ferroptosis, which is negatively regulated by FSP1 activity. FSP1 overexpression can prevent ferroptosis and alleviate emphysema. Next, we found that decreased FSP1 expression was caused by increased N6-methyladenosine modification of FSP1 mRNA. Moreover, the level of FSP1 decreased in a YTHDF2-dependent manner. These results indicate that METTL3-induced FSP1 mRNA methylation leading to low FSP1 expression is a potential therapeutic target for COPD.
Collapse
Affiliation(s)
- Yue Yang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Weiyu Shen
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Zheming Zhang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Youai Dai
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Zixiao Zhang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Tingting Liu
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Jinyan Yu
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Shulun Huang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Yu Ding
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Rong You
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Ziteng Wang
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Yan Wu
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| | - Tao Bian
- Department of Respiratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, People's Republic of China
| |
Collapse
|
23
|
Eggenhofer E, Proneth B. Ferroptosis Inhibition: A Key Opportunity for the Treatment of Ischemia/Reperfusion Injury in Liver Transplantation. Transplantation 2025; 109:e228-e236. [PMID: 39294870 DOI: 10.1097/tp.0000000000005199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
The outcome after liver transplantation has improved in recent years, which can be attributed to superior storage and transportation conditions of the organs, as well as better peri- and postoperative management and advancements in surgical techniques. Nevertheless, there is an increasing discrepancy between the need for organs and their availability. Consequently, the mortality rate on the waiting list is high and continues to rise. One way of counteracting this trend is to increase the use of "expanded criteria donors." This means that more and more donors will be included, especially those who are older and having additional comorbidities (eg, steatosis). A major complication of any transplantation is the occurrence of ischemia/reperfusion injury (IRI), which often leads to liver dysfunction and failure. However, there have been various promising approaches to minimize IRI in recent years, but an effective and clinically applicable method to achieve a better outcome for patients after liver transplantation is still missing. Thereby, the so-called marginal organs are predominantly affected by IRI; thus, it is crucial to develop suitable and effective treatment options for patients. Recently, regulated cell death mechanisms, particularly ferroptosis, have been implicated to play a major role in IRI, including the liver. Therefore, inhibiting this kind of cell death modality presents a promising therapeutic approach for the management of this yet untreatable condition. Thus, this review provides an overview of the role of ferroptosis in liver IRI and transplantation and discusses possible therapeutic solutions based on ferroptosis inhibition to restrain IRI in marginal organs (especially steatosis and donation after circulatory death organs).
Collapse
Affiliation(s)
- Elke Eggenhofer
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Munich, Neuherberg, Germany
| |
Collapse
|
24
|
Wang D, Qu X, Zhang Z, Zhou G. New developments in the role of ferroptosis in sepsis‑induced cardiomyopathy (Review). Mol Med Rep 2025; 31:118. [PMID: 40052561 PMCID: PMC11904766 DOI: 10.3892/mmr.2025.13483] [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: 09/25/2024] [Accepted: 01/15/2025] [Indexed: 03/15/2025] Open
Abstract
Sepsis is a life‑threatening organ dysfunction disorder caused by dysfunctional host response to infection. Sepsis‑induced cardiomyopathy (SIC) is a common and serious complication of sepsis, and it is associated with increased mortality rates; however, its specific pathogenesis is still unclear. Ferroptosis, which is an iron‑dependent form of programmed cell death, is involved in the pathophysiology of SIC. Further study on the mechanism and therapeutic targets of ferroptosis in SIC may provide new strategies for clinical diagnosis and treatment of this condition. The present article reviews the mechanisms between SIC and ferroptosis, summarizes the progress in research of the involvement of ferroptosis in SIC and provides new potential strategies for further research and treatment in the future.
Collapse
Affiliation(s)
- Dingdeng Wang
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Xinguang Qu
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Zhaohui Zhang
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Gaosheng Zhou
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| |
Collapse
|
25
|
Zhao Y, Wang XQ, Liu RQ, Jiang FW, Wang JX, Chen MS, Zhang H, Cui JG, Chang YH, Li JL. SLC7A11 as a therapeutic target to attenuate phthalates-driven testosterone level decline in mice. J Adv Res 2025; 71:369-381. [PMID: 38797476 DOI: 10.1016/j.jare.2024.05.026] [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: 02/16/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024] Open
Abstract
INTRODUCTION Phthalates exposure is a major public health concern due to the accumulation in the environment and associated with levels of testosterone reduction, leading to adverse pregnancy outcomes. However, the relationship between phthalate-induced testosterone level decline and ferroptosis remains poorly defined. OBJECTIVES Herein, we aimed to explore the mechanisms of phthalates-induced testosterone synthesis disorder and its relationship to ferroptosis. METHODS We conducted validated experiments in vivo male mice model and in vitro mouse Leydig TM3 cell line, followed by RNA sequencing and metabolomic analysis. We evaluated the levels of testosterone synthesis-associated enzymes and ferroptosis-related indicators by using qRT-PCR and Western blotting. Then, we analyzed the lipid peroxidation, ROS, Fe2+ levels and glutathione system to confirm the occurrence of ferroptosis. RESULTS In the present study, we used di (2-ethylhexyl) phthalate (DEHP) to identify ferroptosis as the critical contributor to phthalate-induced testosterone level decline. It was demonstrated that DEHP caused glutathione metabolism and steroid synthesis disorders in Leydig cells. As the primary metabolite of DEHP, mono-2-ethylhexyl phthalate (MEHP) triggered testosterone synthesis disorder accompanied by a decrease in the expression of solute carri1er family 7 member 11 (SLC7A11) protein. Furthermore, MEHP synergistically induced ferroptosis with Erastin through the increase of intracellular and mitochondrial ROS, and lipid peroxidation production. Mechanistically, overexpression of SLC7A11 counteracts the synergistic effect of co-exposure to MEHP-Erastin. CONCLUSION Our research results suggest that MEHP does not induce ferroptosis but synergizes Erastin-induced ferroptosis. These findings provide evidence for the role of ferroptosis in phthalates-induced testosterone synthesis disorder and point to SLC7A11 as a potential target for male reproductive diseases. This study established a correlation between ferroptosis and phthalates cytotoxicity, providing a novel view point for mitigating the issue of male reproductive disease and "The Global Plastic Toxicity Debt".
Collapse
Affiliation(s)
- Yi Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xue-Qi Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Rui-Qi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Fu-Wei Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jia-Xin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Ming-Shan Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jia-Gen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yuan-Hang Chang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
26
|
Guo S, Zhang Q, Ge H, Wang H. Baicalin plays a protective role by regulating ferroptosis in multiple diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4837-4849. [PMID: 39661143 DOI: 10.1007/s00210-024-03704-5] [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: 07/27/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
Ferroptosis is a new kind of cell death discovered in recent years, usually accompanied by a large number of lipid peroxidation and iron accumulation in the process of cell death. Ferroptosis has been proven to play an important role in various diseases, including ischemic reperfusion injury, cancer, and neurodegeneration. Therefore, the regulation of ferroptosis will have a vital impact on the occurrence and development of diseases. Baicalin is a flavonoid compound extracted and isolated from the dried roots of Scutellaria baicalensis Georgi, a plant in the family Lamiaceae. It has various biological activities such as antioxidant, anti-proliferative, anti-inflammatory, anti-thrombotic, and regulates apoptosis and ferroptosis. Recently, increasing evidence indicates that baicalin regulation of ferroptosis is involved in multiple diseases. However, the relevant mechanisms are not yet fully understood. Here, we summarized the role of baicalin regulation of ferroptosis in different kinds of diseases, and conducted an in-depth analysis of the relevant mechanisms, hoping to provide the theoretical references for future related researches.
Collapse
Affiliation(s)
- Shiyun Guo
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Qi Zhang
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Hangwei Ge
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
27
|
Qiu Y, Hüther JA, Wank B, Rath A, Tykwe R, Aldrovandi M, Henkelmann B, Mergner J, Nakamura T, Laschat S, Conrad M, Stöhr D, Rehm M. Interplay of ferroptotic and apoptotic cell death and its modulation by BH3-mimetics. Cell Death Differ 2025:10.1038/s41418-025-01514-7. [PMID: 40301648 DOI: 10.1038/s41418-025-01514-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025] Open
Abstract
Ferroptosis and apoptosis are widely considered to be independent cell death modalities. Ferroptotic cell death is a consequence of insufficient radical detoxification and progressive lipid peroxidation, which is counteracted by glutathione peroxidase-4 (GPX4). Apoptotic cell death can be triggered by a wide variety of stresses, including oxygen radicals, and can be suppressed by anti-apoptotic members of the BCL-2 protein family. Mitochondria are the main interaction site of BCL-2 family members and likewise a major source of oxygen radical stress. We therefore studied if ferroptosis and apoptosis might intersect and possibly interfere with one another. Indeed, cells dying from impaired GPX4 activity displayed hallmarks of both ferroptotic and apoptotic cell death, with the latter including (transient) membrane blebbing, submaximal cytochrome-c release and caspase activation. Targeting BCL-2, MCL-1 or BCL-XL with BH3-mimetics under conditions of moderate ferroptotic stress in many cases synergistically enhanced overall cell death and frequently skewed primarily ferroptotic into apoptotic outcomes. Surprisingly though, in other cases BH3-mimetics, most notably the BCL-XL inhibitor WEHI-539, counter-intuitively suppressed cell death and promoted cell survival following GPX4 inhibition. Further studies revealed that most BH3-mimetics possess previously undescribed antioxidant activities that counteract ferroptotic cell death at commonly employed concentration ranges. Our results therefore show that ferroptosis and apoptosis can intersect. We also show that combining ferroptotic stress with BH3-mimetics, context-dependently can either enhance and convert cell death outcomes between ferroptosis and apoptosis or can also suppress cell death by intrinsic antioxidant activities.
Collapse
Affiliation(s)
- Yun Qiu
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Juliana A Hüther
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Bianca Wank
- Institute of Organic Chemistry, University of Stuttgart, Stuttgart, Germany
| | - Antonia Rath
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - René Tykwe
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Maceler Aldrovandi
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Bernhard Henkelmann
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at MRI, TUM, Munich, Germany
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Sabine Laschat
- Institute of Organic Chemistry, University of Stuttgart, Stuttgart, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Daniela Stöhr
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
28
|
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
|
29
|
Zheng J, Zhang W, Ito J, Henkelmann B, Xu C, Mishima E, Conrad M. N-acetyl-l-cysteine averts ferroptosis by fostering glutathione peroxidase 4. Cell Chem Biol 2025:S2451-9456(25)00100-X. [PMID: 40311609 DOI: 10.1016/j.chembiol.2025.04.002] [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: 10/02/2024] [Revised: 03/08/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
N-acetyl-l-cysteine (NAC) is a medication and a widely used antioxidant in cell death research. Despite its somewhat obscure mechanism of action, its role in inhibiting ferroptosis is gaining increasing recognition. In this study, we demonstrate that NAC treatment rapidly replenishes the intracellular cysteine pool, reinforcing its function as a prodrug for cysteine. Interestingly, its enantiomer, N-acetyl-d-cysteine (d-NAC), which cannot be converted into cysteine, also exhibits a strong anti-ferroptotic effect. We further clarify that NAC, d-NAC, and cysteine all act as direct reducing substrates for GPX4, counteracting lipid peroxidation. Consequently, only GPX4-rather than system xc-, glutathione biosynthesis, or ferroptosis suppressor protein 1-is necessary for NAC and d-NAC to prevent ferroptosis. Additionally, we identify a broad range of reducing substrates for GPX4 in vitro, including β-mercaptoethanol. These findings provide new insights into the mechanisms underlying the protective effects of NAC and other potential GPX4-reducing substrates against ferroptosis.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Weijia Zhang
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Junya Ito
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Laboratory of Food Function Analysis, Tohoku University Graduate School of Agricultural Science, Sendai, Miyagi 980-8572, Japan
| | - Bernhard Henkelmann
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Chenxi Xu
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Eikan Mishima
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8574, Japan
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, 85748 Garching, Germany.
| |
Collapse
|
30
|
Ma C, Hu H, Liu H, Zhong C, Wu B, Lv C, Tian Y. Lipotoxicity, lipid peroxidation and ferroptosis: a dilemma in cancer therapy. Cell Biol Toxicol 2025; 41:75. [PMID: 40285867 PMCID: PMC12033115 DOI: 10.1007/s10565-025-10025-7] [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: 11/27/2024] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
The vulnerability of tumor cells to lipid peroxidation, driven by redox imbalance and lipid overabundance within the tumor microenvironment (TME), has become a focal point for novel antitumor strategies. Ferroptosis, a form of regulated cell death predicated on lipid peroxidation, is emerging as a promising approach. Beyond their role in directly eliminating tumor cells, lipid peroxidation and its products, such as 4-hydroxynonenal (HNE), exert an additional influence by damaging DNA and shaping an environment conducive to tumor growth and metastasis. This process polarizes macrophages towards a pro-inflammatory phenotype, dampens the antigen-presenting capacity of dendritic cells (DCs), and undermines the cytotoxic functions of T and NK cells. Furthermore, it transforms neutrophils into pro-tumorigenic polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). The lipid peroxidation of stroma cells also contributes to tumor progression. Although advanced nanotherapies have shown the ability to target tumor cells precisely, they often overlook the nuanced effects of lipid peroxidation products. In this review, we highlight a synergistic mechanism in which lipid peroxidation products and ferroptosis contribute to an immunosuppressive state that is temporally distinct from cell death. This insight broadens our understanding of ferroptosis-derived immunosuppression, encompassing all types of immune cells within the TME. This review aims to catalyze further research in this underexplored area, emphasizing the potential of lipid peroxidation products to hinder the clinical translation of ferroptosis-based therapies.
Collapse
Affiliation(s)
- Chuhan Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Huixin Hu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Hao Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Chongli Zhong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Baokang Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Chao Lv
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
31
|
Yang J, Shi X, Ma M, Li Z, Liu H, Cui Y, Xu Z, Wang J. PGC-1α role in rescuing ferroptosis in cerebral ischemia/reperfusion injury through promoting mitochondrial biogenesis and UCP2 expression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167874. [PMID: 40294850 DOI: 10.1016/j.bbadis.2025.167874] [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: 12/01/2024] [Revised: 04/02/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
Cerebral ischemia/reperfusion injury (CIRI) is a critical factor leading to adverse outcomes in acute ischemic stroke with reperfusion therapy. The occurrence of CIRI involves several cell death pathways, such as ferroptosis. Peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) a vital role in mitochondrial biogenesis and induces several crucial reactive oxygen species (ROS) detoxifying enzymes. Nonetheless, the role of activated PGC-1α in CIRI is still unclear. In this research, we utilized a PGC-1α agonist (ZLN005) in both in vitro and in vivo models of CIRI and found that ZLN005 ameliorates neurologic deficits, reduces infarct volume, and inhibits neuronal ferroptosis in CIRI. Furthermore, CIRI led to a decrease in neuronal mitochondrial quantity and downregulation of uncoupling protein 2 (UCP2) expression. Treatment with ZLN005 activated PGC-1α, promoted neuronal mitochondrial biogenesis, and upregulated UCP2 expression, thereby reducing mitochondrial oxidative stress. The application of the mitochondria-targeted antioxidant Mito-TEMPO inhibited ferroptosis, while UCP2 silencing induced mitochondrial oxidative stress and weakened ZLN005 inhibitory effect of ferroptosis, confirming the dependency of ferroptosis on mitochondrial oxidative stress in CIRI. According to these findings, targeting PGC-1α may offer an effective therapeutic strategy for CIRI by regulating mitochondrial homeostasis and protecting neurons from ferroptotic damage.
Collapse
Affiliation(s)
- Jiahui Yang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Xiaohua Shi
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Ming Ma
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Zheng Li
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Hongyu Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Yang Cui
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Zhongxin Xu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China.
| | - Jiaoqi Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
32
|
Zhou P, Liu M, Lv T. Ferroptosis targeting offers a therapeutic target for septic cardiomyopathy. Tissue Cell 2025; 95:102930. [PMID: 40288080 DOI: 10.1016/j.tice.2025.102930] [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: 01/23/2025] [Revised: 03/05/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Sepsis-induced cardiac dysfunction, usually termed sepsis-induced cardiomyopathy or septic cardiomyopathy(SCM), is developed in approximately 70 % of the patients with sepsis, making it is a major concern for sepsis patients. However, the pathogenesis of SCM remain incompletely understood. Ferroptosis, a newly identified mechanism of regulated cell death, characterized by a decline in antioxidant capacity, iron accumulation, and lipid peroxidation(LPO), is involved in sepsis and SCM. Moreover, ferroptosis inhibitors confer a novel therapeutic regimen in SCM. In this Review, we first summarizes the core mechanism of ferroptosis, with an emphasis on how best to interpret ferroptosis leads to the genesis of SCM. We then highlights our focus on the emerging different types of therapeutic ferroptosis inhibitors and summarizes their pharmacological beneficial effect to treat SCM. This review highlights a novel potential therapeutic strategy for SCM by pharmacologically inhibiting ferroptosis.
Collapse
Affiliation(s)
- Pengsi Zhou
- Department of Cardiology, The Affiliated Hospital of Chifeng University, Chifeng 024005, China.
| | - Mengxue Liu
- Department of Cardiology, The Affiliated Hospital of Chifeng University, Chifeng 024005, China
| | - Tao Lv
- Department of Cardiology, The Affiliated Hospital of Chifeng University, Chifeng 024005, China.
| |
Collapse
|
33
|
Yang Y, Jiang B, Shi L, Wang L, Yang Y, Li Y, Zhang Y, Zhu Z, Zhang X, Liu X. The potential of natural herbal plants in the treatment and prevention of non-small cell lung cancer: An encounter between ferroptosis and mitophagy. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119555. [PMID: 40015539 DOI: 10.1016/j.jep.2025.119555] [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: 08/24/2024] [Revised: 02/15/2025] [Accepted: 02/22/2025] [Indexed: 03/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine constitutes a substantial cultural and scientific resource for the Chinese nation, attracting considerable scholarly interest due to its intrinsic characteristics of "multi-component, multi-target, and multi-pathway" interactions. Simultaneously, it aligns accurately with the intricate and continuously evolving progression of non-small cell lung cancer (NSCLC). Furthermore, contemporary pharmacological studies indicate that natural herbaceous plants and their bioactive compounds exhibit a diverse array of biological activities, including antioxidant, anti-inflammatory, and anti-tumor effects, among others. Additionally, these substances have been demonstrated to possess a degree of safety, particularly in terms of exhibiting comparatively lower levels of toxicity to the liver and kidneys when contrasted with conventional Western medicine. Thus, the development of herbal plants, which includes both single herbs and composite formulations, as well as their bioactive constituents, through the targeted regulation of ferroptosis and mitophagy, presents substantial potential and instills considerable hope for individuals diagnosed with NSCLC. AIM OF THE REVIEW This review aims to conduct a critical analysis of the ethnopharmacological applications of natural herbaceous plants in relation to ferroptosis and mitophagy in NSCLC. The objective is to evaluate the potential advantages of prioritizing specific phytochemical constituents found in these plants, which may serve as novel therapeutic candidates informed by ethnobotanical knowledge. Additionally, this study seeks to enhance the current pharmacological applications of natural herbaceous plants. METHODS An investigation into natural herbal remedies for NSCLC was conducted, with a particular emphasis on the ferroptosis and mitophagy pathways. This study utilized traditional medical texts and ethnomedicinal literature as primary sources. Furthermore, relevant information related to ethnobotany, phytochemistry, and pharmacology is obtained from online databases, including PubMed and the China National Knowledge Infrastructure (CNKI), among others. "Traditional Chinese medicine compound preparations", "single herb extracts", "active compounds", "NSCLC", "ferroptosis", and "mitophagy" were used as keywords when searching the databases. Consequently, pertinent articles published in recent years were collected and analyzed. RESULTS Given the complex etiology of NSCLC, treatment strategies that concentrate exclusively on ferroptosis or mitophagy often demonstrate limitations. In this regard, the utilization of herbal plants offers unique benefits in the management of NSCLC. The rationale can be summarized within the following two dimensions: Firstly, due to the molecular mechanisms of ferroptosis and mitophagy involving multiple signaling pathways (including PINK1/Parkin, HMGB1, system Xc-/GPX4/GSH, FSP1/CoQ10/NAD (P) H, and so on), sometimes drugs with a single target are difficult to involve multiple pathways. Fortunately, there is an expanding body of evidence suggesting that various herbaceous plants and their bioactive compounds can affect multiple biological targets. Moreover, these compounds seem to interact with several targets associated with ferroptosis and mitophagy in NSCLC (such as NIX, BNIP3, FUNDC1, GPX4, FSP1, P53, Nrf2, LncRNA, and so on). Secondly, Herbaceous plants and their bioactive compounds have been shown to possess a favorable safety profile, particularly with respect to reduced hepatotoxicity and nephrotoxicity in comparison to conventional Western medicine. For example, Numerous compound formulations, such as Fangji Huangqi decoction, Mufangji decoction, Qiyu Sanlong decoction, and Fuzheng Kangai decoction, have been employed in China for millennia, and their clinical efficacy appears to be quite promising. Notably, In recent years, numerous researchers have sought to isolate active constituents from clinically effective compound formulations through the application of chemical methodologies. This endeavor has been driven by the necessity to tackle challenges related to complex ingredient compositions and sophisticated processing. These active compounds have been employed in cellular and animal studies to elucidate the molecular mechanisms underlying these formulations. CONCLUSIONS The Asian region has a long-standing historical tradition of employing natural herbaceous plants for traditional medicinal purposes. Phytochemical and pharmacological studies have shown that various compound preparations derived from traditional Chinese medicine, along with individual herb extracts and their active constituents, display a range of bioactive effects. These effects encompass anti-tumor, anti-inflammatory, antibacterial, and antioxidant properties, among others. Numerous traditional compound formulations originating from China have emerged as promising candidates for the development of pharmacological agents targeting NSCLC. It is noteworthy that a variety of compound formulations aimed at the ferroptosis and mitophagy pathways, which demonstrate unique therapeutic effects on NSCLC, are presently under extensive investigation by an increasing number of researchers. Therefore, it is imperative to consider in vitro mechanistic studies, in vivo pharmacological evaluations, and assessments of clinical efficacy. Furthermore, it is essential to conduct a comprehensive assessment of plant resources, implement quality control measures, and engage in toxicological research to ensure that the data is appropriate for further examination.
Collapse
Affiliation(s)
- Yujie Yang
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Lijuan Shi
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Lili Wang
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Yaru Yang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Yongyu Li
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Yanmei Zhang
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Zhongbo Zhu
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Xuhui Zhang
- Department of Pulmonary Diseases, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, 730030, China.
| | - Xiping Liu
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, Gansu, 730000, China; Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, Gansu, 730000, China; Department of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
34
|
Xu J, Shen R, Qian M, Zhou Z, Xie B, Jiang Y, Yu Y, Dong W. Ferroptosis in Alzheimer's Disease: The Regulatory Role of Glial Cells. J Integr Neurosci 2025; 24:25845. [PMID: 40302253 DOI: 10.31083/jin25845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/11/2024] [Accepted: 10/30/2024] [Indexed: 05/02/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by the formation of amyloid plaques, neurofibrillary tangles and progressive cognitive decline. Amyloid-beta peptide (Aβ) monoclonal antibody therapeutic clinical trials have nearly failed, raising significant concerns about other etiological hypotheses about AD. Recent evidence suggests that AD patients also exhibit persistent neuronal loss and neuronal death accompanied by brain iron deposition or overload-related oxidative stress. Ferroptosis is a type of cell death that depends on iron, unlike autophagy and apoptosis. Inhibiting neuronal ferroptosis function is effective in improving cognitive impairment in AD. Notably, new research shows that ferroptosis in AD is crucially dependent on glial cell activation. This review examines the relationship between the imbalance of iron metabolism, the regulation of iron homeostasis in glial cells and neuronal death in AD pathology. Finally, the review summarizes some current drug research in AD targeting iron homeostasis, many novel iron-chelating compounds and natural compounds showing potential AD-modifying properties that may provide therapeutic targets for treating AD.
Collapse
Affiliation(s)
- Jingyi Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Rongjing Shen
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Mengting Qian
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Zhengjun Zhou
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, Institute of Epigenetics and Brain Science, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yong Jiang
- Laboratory of Neurological Diseases and Brain Function, Institute of Epigenetics and Brain Science, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yang Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
35
|
Gujar V, Li H, Paull TT, Neumann CA, Weyemi U. Unraveling the nexus: Genomic instability and metabolism in cancer. Cell Rep 2025; 44:115540. [PMID: 40208791 PMCID: PMC12043202 DOI: 10.1016/j.celrep.2025.115540] [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: 11/25/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025] Open
Abstract
The DNA-damage response (DDR) is a signaling network that enables cells to detect and repair genomic damage. Over the past three decades, inhibiting DDR has proven to be an effective cancer therapeutic strategy. Although cancer drugs targeting DDR have received approval for treating various cancers, tumor cells often develop resistance to these therapies, owing to their ability to undergo energetic metabolic reprogramming. Metabolic intermediates also influence tumor cells' ability to sense oxidative stress, leading to impaired redox metabolism, thus creating redox vulnerabilities. In this review, we summarize recent advances in understanding the crosstalk between DDR and metabolism. We discuss combination therapies that target DDR, metabolism, and redox vulnerabilities in cancer. We also outline potential obstacles in targeting metabolism and propose strategies to overcome these challenges.
Collapse
Affiliation(s)
- Vaibhavi Gujar
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haojian Li
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tanya T Paull
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Carola A Neumann
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Urbain Weyemi
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
36
|
Zhang S, Xiao H, Lin Y, Tang X, Tong W, Shao B, Li H, Xu L, Ding X, Chai R. Targeting Programmed Cell Death in Acquired Sensorineural Hearing Loss: Ferroptosis, Necroptosis, and Pyroptosis. Neurosci Bull 2025:10.1007/s12264-025-01370-y. [PMID: 40261527 DOI: 10.1007/s12264-025-01370-y] [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: 10/23/2024] [Accepted: 12/06/2024] [Indexed: 04/24/2025] Open
Abstract
Sensorineural hearing loss (SNHL), the most commonly-occurring form of hearing loss, is caused mainly by injury to or the loss of hair cells and spiral ganglion neurons in the cochlea. Numerous environmental and physiological factors have been shown to cause acquired SNHL, such as ototoxic drugs, noise exposure, aging, infections, and diseases. Several programmed cell death (PCD) pathways have been reported to be involved in SNHL, especially some novel PCD pathways that have only recently been reported, such as ferroptosis, necroptosis, and pyroptosis. Here we summarize these PCD pathways and their roles and mechanisms in SNHL, aiming to provide new insights and potential therapeutic strategies for SNHL by targeting these PCD pathways.
Collapse
Affiliation(s)
- Shasha Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology-Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China.
| | - Hairong Xiao
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology-Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China
| | - Yanqin Lin
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology-Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China
| | - Xujun Tang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology-Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Wei Tong
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology-Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Buwei Shao
- School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - He Li
- Department of Otolaryngology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China.
| | - Xiaoqiong Ding
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology-Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology-Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
- Department of Neurology, Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Institute for Stem Cells and Regeneration, Chinese Academy of Science, Beijing, 100081, China.
| |
Collapse
|
37
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
38
|
Singh M, Arora HL, Naik R, Joshi S, Sonawane K, Sharma NK, Sinha BK. Ferroptosis in Cancer: Mechanism and Therapeutic Potential. Int J Mol Sci 2025; 26:3852. [PMID: 40332483 DOI: 10.3390/ijms26083852] [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: 02/11/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer drug resistance occurs when cancer cells evade cell death following treatment with chemotherapy, radiation therapy, and targeted therapies. This resistance is often linked to the reprogramming of programmed cell death (PCD) pathways, allowing cancer cells to survive drug-induced stress. However, certain anticancer therapies, when combined with specific agents or inhibitors, can induce ferroptosis-a form of cell death driven by iron-dependent lipid peroxidation. Currently, extensive preclinical and clinical research is underway to investigate the molecular, cellular, and tissue-specific mechanisms underlying ferroptosis, with the goal of identifying strategies to overcome drug resistance in cancers unresponsive to conventional PCD pathways. By harnessing ferroptosis, cancer cells can be compelled to undergo lipid peroxidation-induced death, potentially improving therapeutic outcomes in patients with cancer. This short review aims to enhance the understanding of ferroptosis inducers in cancer therapy and stimulate further research into ferroptosis-based approaches for more effective clinical cancer treatment.
Collapse
Affiliation(s)
- Mansaa Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India
| | - Hasmiq L Arora
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India
| | - Rutuja Naik
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India
| | - Shravani Joshi
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India
| | - Kaveri Sonawane
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India
| | - Birandra K Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
39
|
Saeed BI, Uthirapathy S, Kubaev A, Ganesan S, Shankhyan A, Gupta S, Joshi KK, Kariem M, Jasim AS, Ahmed JK. Ferroptosis as a key player in the pathogenesis and intervention therapy in liver injury: focusing on drug-induced hepatotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04115-w. [PMID: 40244448 DOI: 10.1007/s00210-025-04115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Globally, drug-induced hepatotoxicity or drug-induced liver injury (DILI) is a serious clinical concern. Knowing the processes and patterns of cell death is essential for finding new therapeutic targets since there are not many alternatives to therapy for severe liver lesions. Excessive lipid peroxidation is a hallmark of ferroptosis, an iron-reliant non-apoptotic cell death linked to various liver pathologies. When iron is pathogenic, concomitant inflammation may exacerbate iron-mediated liver injury, and the hepatocyte necrosis that results is a key element in the fibrogenic response. The idea that dysregulated metabolic pathways and compromised iron homeostasis contribute to the development of liver injury by ferroptosis is being supported by new data. Various ferroptosis-linked genes and pathways have been linked to liver injury, although the molecular processes behind ferroptosis's pathogenicity are not well known. Here, we delve into the features of ferroptosis, the processes governing ferroptosis, and our current knowledge of iron metabolism. We also provide an overview of ferroptosis's involvement in the pathophysiology of liver injury, particularly DILI. Lastly, the therapeutic possibilities of ferroptosis targeting for liver injury management have been provided. Natural products, nanoparticles (NPs), mesenchymal stem cell (MSC), and their exosomes have attracted increasing attention among such therapeutics.
Collapse
Affiliation(s)
- Bahaa Ibrahim Saeed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Jawad Kadhim Ahmed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
40
|
Jiang Y, Saeed TN, Alfarttoosi KH, Bishoyi AK, Rekha MM, Kundlas M, Jain B, Rizaev J, Taher WM, Alwan M, Jawad MJ, Ali Al-Nuaimi AM. The intersection of ferroptosis and non-coding RNAs: a novel approach to ovarian cancer. Eur J Med Res 2025; 30:300. [PMID: 40247379 PMCID: PMC12007203 DOI: 10.1186/s40001-025-02559-7] [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: 02/08/2025] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
Understanding the core principles of ovarian cancer has been significantly improved through the exploration of Ferroptosis, a type of cell death triggered by iron that leads to an increase in lipid peroxides. Current research has shed light on the critical functions of non-coding RNAs, such as circRNAs, lncRNAs, and miRNAs, in regulating ferroptosis in ovarian cancer. The aim of this paper is to comprehensively analyze how ncRNAs influence the development of ferroptosis in ovarian cancer cells. In-depth exploration is undertaken to understand the intricate ways in which ncRNAs regulate essential elements of ferroptosis, including iron management and lipid peroxidation levels. We also investigate their significant involvement in the progression of this type of cellular demise. It should be emphasized that ncRNAs can impact the synthesis of crucial proteins, such as GPX4, a key contributor to the cellular defense against oxidation, and ACSL4, involved in lipid formation. In addition, we examine the correlation between ncRNAs and well-known pathways associated with oxidative stress and cell death. The consequences of these discoveries are noteworthy, since focusing on particular ncRNAs could potentially render ovarian cancer cells more vulnerable to ferroptosis, effectively combating drug resistance problems. This discussion highlights the growing significance of ncRNAs in governing ferroptosis and their potential as useful biomarkers and treatment targets for ovarian cancer. We intend to promote additional research into the involvement of ncRNAs in controlling ferroptosis, based on current findings, with the ultimate goal of informing targeted therapeutic strategies and improving long-term treatment outcomes for individuals suffering from OC.
Collapse
Affiliation(s)
- Youyi Jiang
- School of Civil Engineering, Chongqing Jiaotong University, Chongqing, China
| | - Tamara Nazar Saeed
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq.
| | | | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Bhavik Jain
- Chitkara Centre for Research and Development, Chitkara University, Baddi, Himachal Pradesh, 174103, India
| | - Jasur Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
41
|
Zhao N, Li S, Wu H, Wei D, Pu N, Wang K, Liu Y, Tao Y, Song Z. Ferroptosis: An Energetic Villain of Age-Related Macular Degeneration. Biomedicines 2025; 13:986. [PMID: 40299661 PMCID: PMC12024642 DOI: 10.3390/biomedicines13040986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Iron homeostasis plays an important role in maintaining cellular homeostasis; however, excessive iron can promote the production of reactive oxygen species (ROS). Ferroptosis is iron-dependent programmed cell death that is characterized by excessive iron accumulation, elevated lipid peroxides, and the overproduction of ROS. The maintenance of iron homeostasis is contingent upon the activity of the transferrin receptor (TfR), ferritin (Ft), and ferroportin (FPn). In the retina, iron accumulation and lipid peroxidation can contribute to the development of age-related macular degeneration (AMD). This phenomenon can be explained by the occurrence of the Fenton reaction, in which the interaction between divalent iron and hydrogen peroxide leads to the generation of highly reactive hydroxyl radicals. The hydroxyl radicals exhibit a propensity to attack proteins, lipids, nucleic acids, and carbohydrates, thereby instigating oxidative damage and promoting lipid peroxidation. Ultimately, these processes culminate in cell death and retinal degeneration. In this context, a comprehensive understanding of the exact mechanisms underlying ferroptosis may hold significant importance for developing therapeutic interventions. This review summarizes recent findings on iron metabolism, cellular ferroptosis, and lipid metabolism in the aging retina. We also introduce developments in the therapeutic strategies using iron chelating agents. Further refinements of these knowledges would deepen our comprehension of the pathophysiology of AMD and advance the clinical management of degenerative retinopathy. A comprehensive search strategy was employed to identify relevant studies on the role of ferroptosis in AMD. We performed systematic searches of the PubMed and Web of Science electronic databases from inception to the current date. The keywords used in the search included "ferroptosis", "AMD", "age-related macular degeneration", "iron metabolism", "oxidative stress", and "ferroptosis pathways". Peer-reviewed articles, including original research, reviews, meta-analyses, and clinical studies, were included in this paper, with a focus on the molecular mechanisms of ferroptosis in AMDs. Studies not directly related to ferroptosis, iron metabolism, or oxidative stress in the context of AMD were excluded. Furthermore, articles that lacked sufficient data or were not peer-reviewed (e.g., conference abstracts, editorials, or opinion pieces) were not considered.
Collapse
Affiliation(s)
- Na Zhao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Siyu Li
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Hao Wu
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Dong Wei
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Ning Pu
- College of Medicine, Zhengzhou University, Zhengzhou 450001, China; (S.L.); (H.W.); (D.W.); (N.P.)
| | - Kexin Wang
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Yashuang Liu
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Ye Tao
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| | - Zongming Song
- Henan Eye Institute, Henan Eye Hospital, People’s Hospital of Zhengzhou University, Henan University School of Medicine, Henan Provincial People’s Hospital, Zhengzhou 450003, China; (N.Z.); (K.W.); (Y.L.)
| |
Collapse
|
42
|
Direksunthorn T, T Ahmed A, Pluetrattanabha N, Uthirapathy S, Ballal S, Singh A, Al-Hetty HRAK, Devi A, Sharma GC, Yumashev A. Ferroptosis in immune chaos: Unraveling its impact on disease and therapeutic potential. J Physiol Biochem 2025:10.1007/s13105-025-01078-7. [PMID: 40237936 DOI: 10.1007/s13105-025-01078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
Since its introduction in 2012, ferroptosis has garnered significant attention from researchers over the past decade. Unlike autophagy and apoptosis, ferroptosis is an atypical iron-dependent programmed cell death that falls under necrosis. It is regulated by various cellular metabolic and signaling processes, which encompass amino acid, lipid, iron, and mitochondrial metabolism. The initiation of ferroptosis occurs through iron-dependent phospholipid peroxidation. Notably, ferroptosis exhibits a dual effect and is associated with various diseases. A significant challenge lies in managing autoimmune disorders with unknown origins that stem from the reactivation of the immune system. Two contributing factors to autoimmunity are the aberrant stimulation of cell death and the inadequate clearance of dead cells, which can expose or release intracellular components that activate the immune response. Ferroptosis is distinct from other forms of cell death, such as apoptosis, necroptosis, autophagy, and pyroptosis, due to its unique morphological, biochemical, and genetic characteristics and specific relationship with cellular iron levels. Recent studies indicate that immune cells can both induce and undergo ferroptosis. To better understand how ferroptosis influences immune responses and its imbalance in disease, a molecular understanding of the relationship between ferroptosis and immunity is essential. Consequently, further research is needed to develop immunotherapeutics that target ferroptosis. This review primarily focuses on the role of ferroptosis in immune-related disorders.
Collapse
Affiliation(s)
| | | | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | | | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
43
|
Liu Y, Yu Z, Lu Y, Liu Y, Chen L, Li J. Progress in the study of the mechanism of ferroptosis in coronary heart disease and clinical intervention strategies. Front Cardiovasc Med 2025; 12:1545231. [PMID: 40308274 PMCID: PMC12040834 DOI: 10.3389/fcvm.2025.1545231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Coronary heart disease (CHD), a serious cardiovascular condition with complex and diverse pathogenesis, has recently seen increased attention to the role of ferroptosis-a novel iron-dependent form of programmed cell death. This review synthesizes current research on ferroptosis mechanisms in CHD and emerging clinical intervention strategies. Ferroptosis is characterized by dysregulated iron metabolism, lipid peroxidation, and reactive oxygen species (ROS) accumulation, processes intimately linked to CHD pathophysiology. Under ischemic and hypoxic conditions commonly seen in coronary artery disease (CAD), cardiomyocytes become particularly susceptible to ferroptosis, resulting in cellular dysfunction and diminished cardiac performance. Mechanistic studies have revealed that altered expression of iron metabolism-related proteins (including GPX4, FTH1, TfR1, and HO-1), accumulation of lipid peroxidation products, and disruption of antioxidant defense systems (particularly the Nrf2/GPX4 pathway) are central to ferroptosis progression in cardiac tissue. Clinically, both specific ferroptosis inhibitors (such as Ferrostatin-1) and traditional medicine components (such as Puerarin) have emerged as promising therapeutic candidates, showing cardioprotective effects in experimental models. However, research into ferroptosis mechanisms in CHD remains in its early stages, with significant questions regarding its relationship with other cell death pathways and the clinical efficacy of ferroptosis-targeting interventions requiring further investigation. Future research directions should include in-depth mechanistic exploration and the development of more effective, safer clinical interventions targeting the ferroptosis pathway in cardiovascular disease.
Collapse
Affiliation(s)
- Yingzhi Liu
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Zixuan Yu
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Yuwen Lu
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Yue Liu
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Lingli Chen
- Hunan Key Laboratory of Pathogeny Biology of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jie Li
- Hunan Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
44
|
Saimoto Y, Kusakabe D, Morimoto K, Matsuoka Y, Kozakura E, Kato N, Tsunematsu K, Umeno T, Kiyotani T, Matsumoto S, Tsuji M, Hirayama T, Nagasawa H, Uchida K, Karasawa S, Jutanom M, Yamada KI. Lysosomal lipid peroxidation contributes to ferroptosis induction via lysosomal membrane permeabilization. Nat Commun 2025; 16:3554. [PMID: 40229298 PMCID: PMC11997074 DOI: 10.1038/s41467-025-58909-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
Ferroptosis, a form of cell death instigated by iron-dependent lipid peroxidation reactions (LPO), is emerging as a promising therapeutic target for cancer. While the mechanisms governing LPO induction and suppression have gradually been unveiled, questions persist regarding the specific cellular location of LPO and the utilization of iron in driving cell death. A comprehensive understanding of these aspects holds significant potential for advancing therapeutic applications in disease management. Here, we show lysosomal LPO in the initiation of ferroptosis, leveraging the hidden abilities of fluorescent detection probes. Intra-lysosomal LPO triggers iron leakage, fostering cell-wide LPO by augmenting lysosomal membrane permeabilization (LMP). Conversely, cell lines with low susceptibility to ferroptosis do not exhibit LMP. This deficiency is rectified by the concurrent administration of chloroquine, leading to LMP induction and subsequent cell death. These findings underscore enhancing LMP induction efficacy as a strategic approach to surmount resistance to therapies in cancer.
Collapse
Affiliation(s)
- Yuma Saimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Daiki Kusakabe
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazushi Morimoto
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuta Matsuoka
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Eisho Kozakura
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nao Kato
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kayoko Tsunematsu
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomohiro Umeno
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, 194-8543, Japan
| | - Tamiko Kiyotani
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, 194-8543, Japan
| | - Shota Matsumoto
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, 194-8543, Japan
| | - Mieko Tsuji
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Tasuku Hirayama
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Hideko Nagasawa
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu, 501-1196, Japan
| | - Koji Uchida
- Laboratory of Food Chemistry and Life Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Satoru Karasawa
- Faculty of Pharmaceutical Sciences, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, 194-8543, Japan
| | - Mirinthorn Jutanom
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ken-Ichi Yamada
- Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
45
|
Ruan H, Xiang H, Liu Y, Wang P, Dong L, Cao Y, Liang D, Ding Z. FSP1 regulates ferroptosis and mitochondrial function during mouse oocyte maturation. Exp Cell Res 2025; 447:114524. [PMID: 40113029 DOI: 10.1016/j.yexcr.2025.114524] [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: 02/09/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Oocyte quality plays a fundamental role in fertilization and embryonic development. Emerging evidence indicates that ferroptosis may impair oocyte quality. Ferroptosis suppressor protein 1 (FSP1), a known ferroptosis inhibitor, has an uncharacterized function in regulating oocyte quality during meiotic maturation. This study identified FSP1 expression across all stages of meiotic maturation with localization to the cytoplasm of mouse oocytes. Aged mice exhibited a marked reduction in FSP1 expression within the ovaries and oocytes. Pharmacological inhibition of FSP1 disrupted germinal vesicle breakdown and polar body emission, leading to spindle defects and chromosome misalignment. Additionally, FSP1 inhibition persistently activated the spindle assembly checkpoint, resulting in meiotic arrest. At the mechanistic level, inhibition of FSP1 led to an increase in intracellular Fe2+ levels, enhanced dihydroethidium fluorescence, excessive accumulation of reactive oxygen species, and intensified lipid peroxidation. Disruptions in ferroptosis-associated gene expression further indicated that oocytes underwent ferroptosis. Moreover, mitochondrial dysfunction was evident following FSP1 inhibition, as reflected by aberrant mitochondrial distribution, diminished ATP production, and an elevated mitochondrial membrane potential. Collectively, these results establish FSP1 as a key regulator of oocyte meiotic maturation by modulating iron homeostasis and mitochondrial function, while its inhibition triggers ferroptosis-dependent meiotic failure.
Collapse
Affiliation(s)
- Hongzhen Ruan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China
| | - Huifen Xiang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China
| | - Yajing Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China
| | - Peiwen Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China
| | - Liuliu Dong
- Bengbu Medical University, No. 2600 Donghai Avenue, Bengbu, 233030, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China.
| | - Dan Liang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China.
| | - Zhiming Ding
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No.81 Meishan Road, Hefei, 230032, China; Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Ministry of Education of the People's Republic of China, No.81 Meishan Road, Hefei, 230032, China.
| |
Collapse
|
46
|
Huang C, Zeng Q, Chen J, Wen Q, Jin W, Dai X, Ruan R, Zhong H, Xia Y, Wu Z, Huang R, Zhang J, Yao Y, Li L, Lei W, Xiong J, Deng J. TMEM160 inhibits KEAP1 to suppress ferroptosis and induce chemoresistance in gastric cancer. Cell Death Dis 2025; 16:287. [PMID: 40223081 PMCID: PMC11994801 DOI: 10.1038/s41419-025-07621-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
Chemoresistance is the most significant challenge affecting the clinical efficacy of the treatment of patients with gastric cancer (GC). Here we reported that transmembrane protein 160 (TMEM160) suppressed ferroptosis and induced chemoresistance in GC cells. Mechanistically, TMEM160 recruited the E3 ligase TRIM37 to promote K48-linked ubiquitination and degradation of KEAP1, thereby activating NRF2 and transcriptionally upregulating the target genes GPX4 and SLC7A11 to inhibit ferroptosis. Further in vitro and in vivo experiments demonstrated that the combination of TMEM160 targeting and chemotherapy had a synergistic inhibitory effect on the growth of GC cells, which was partially NRF2-dependent. Moreover, TMEM160 and NRF2 protein levels were markedly overexpressed in GC tissues, and their co-overexpression was an independent factor for poor prognosis. Collectively, these findings indicate that TMEM160, as a pivotal negative regulator of ferroptosis, exerts a crucial influence on the chemoresistance of GC through the TRIM37-KEAP1/NRF2 axis, providing a potential new prognostic factor and combination therapy strategy for patients with GC.
Collapse
Affiliation(s)
- Chunye Huang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Qinru Zeng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Jingyi Chen
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Qin Wen
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Weilun Jin
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Xiaofeng Dai
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Hongguang Zhong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Yang Xia
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Zhipeng Wu
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Ruixuan Huang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Jianxi Zhang
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Li Li
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wan Lei
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, Nanchang, China.
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
47
|
Song W, Sun P, Zhao T, Zang Y, Dong P, Tang Q, Chen W, Chen W, Wang Z, Zhang Q, Wang Y, Yin C, Yu M. Unveiling the therapeutic potential of ferroptosis in lung cancer: a comprehensive bibliometric analysis and future therapeutic insights. Discov Oncol 2025; 16:508. [PMID: 40208519 PMCID: PMC11985706 DOI: 10.1007/s12672-025-02234-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Lung cancer remains the leading cause of cancer-related deaths worldwide, with increasing attention being given to novel therapeutic strategies that target the mechanisms underlying tumor growth and drug resistance. Among these, ferroptosis, a regulated cell death driven by iron-dependent lipid peroxidation, has become a key focus in cancer research. Despite extensive research, the exact role of ferroptosis in lung cancer progression and treatment remains unclear, especially regarding its interaction with immune cells and the tumor microenvironment. OBJECTIVE AND METHODS To address these limitations, this study utilizes a comprehensive bibliometric analysis to explore the current landscape of ferroptosis research in lung cancer. We collected data from the Web of Science Core Collection, covering articles published between 2015 and 2025, and analyzed them using advanced tools such as VOSviewer and CiteSpace. RESULTS This study uses a comprehensive bibliometric analysis to uncover key trends and emerging areas related to lung cancer in ferroptosis research. Recently, the focus has shifted from basic mechanisms to clinical applications, particularly in developing GPX4-targeted therapies and combination treatments. With increasing international collaboration, the United States and China have become key players. Interdisciplinary research, especially on ferroptosis and the cancer-immune system, offers new insights into its role in the tumor microenvironment and immunotherapy. Ferroptosis shows excellent promise in overcoming drug resistance by regulating iron-dependent lipid peroxidation and enhancing treatment efficacy. Future research should focus on ferroptosis' clinical translation, particularly in personalized medicine and overcoming resistance, offering broad prospects for lung cancer treatment. CONCLUSION This paper provides valuable insights into the trends, key contributors, and emerging frontiers of ferroptosis research in lung cancer. It identifies important developments that can serve as a foundation for translating ferroptosis-based therapies into clinical practice, particularly to address drug resistance in lung cancer.
Collapse
Affiliation(s)
- Wenhuan Song
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
- Binzhou Medical University, Binzhou, China
| | - Peipei Sun
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Tongzhen Zhao
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Yunxue Zang
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Pengpeng Dong
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Qi Tang
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Wenyu Chen
- Binzhou Medical University, Binzhou, China
| | - Wenyi Chen
- Binzhou Medical University, Binzhou, China
| | | | | | | | - Chunhui Yin
- Shandong Academy of Medical Sciences (SDAMS), Jinan, China
| | - Mingkun Yu
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China.
- Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
48
|
Wang X, Xue Y, Chang L, Zhu X, Liu W, Liang T. The Regulation of Trace Metal Elements in Cancer Ferroptosis. Adv Biol (Weinh) 2025:e2400821. [PMID: 40200790 DOI: 10.1002/adbi.202400821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/11/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, as novel type of regulated cell death that has garnered widespread attention over the past decade, has witnessed the continuous discovery of an increasing number of regulatory mechanisms. Trace metal elements play a multifaceted and crucial role in oncology. Interestingly, it has been increasingly evident that these elements, such as copper, are involved in the regulation of iron accumulation, lipid peroxidation and antiferroptotic systems, suggesting the existence of "nonferrous" mechanisms in ferroptosis. In this review, a comprehensive overview of the composition and mechanism of ferroptosis is provided. The interaction between copper metabolism (including cuproptosis) and ferroptosis in cancer, as well as the roles of other trace metal elements (such as zinc, manganese, cobalt, and molybdenum) in ferroptosis are specifically focused. Furthermore, the applications of nanomaterials based on these metals in cancer therapy are also reviewed and potential strategies for co-targeting ferroptosis and cuproptosis are explored. Nevertheless, in light of the intricate and ambiguous nature of these interactions, ongoing research is essential to further elucidate the "nonferrous" mechanisms of ferroptosis, thereby facilitating the development of novel therapeutic targets and approaches for cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuanyuan Xue
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lei Chang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xuena Zhu
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Wenjun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
- Zhejiang University Cancer Center, Hangzhou, 310003, China
| |
Collapse
|
49
|
Du H, Yang K, Yang J, Wan J, Pan Y, Song W, Xu S, Chen C, Li J. Euphorbia humifusa Willd. ex Schltdl. Mitigates Liver Injury via KEAP1-NFE2L2-Mediated Ferroptosis Regulation: Network Pharmacology and Experimental Validation. Vet Sci 2025; 12:350. [PMID: 40284852 PMCID: PMC12030869 DOI: 10.3390/vetsci12040350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Liver injury poses major health risks in livestock, necessitating effective therapeutic interventions. This study elucidates the hepatoprotective mechanisms of Euphorbia humifusa Willd. ex Schltdl. (EHW) by integrating network pharmacology, molecular docking, and experimental validation. Using a CCl4-induced liver injury model mimicking veterinary clinical scenarios, EHW markedly alleviated hepatic damage, demonstrated by reduced liver index, serum ALT and AST levels, histopathological lesions, iron accumulation, inflammatory cytokines, and ferroptosis-associated gene expression. Network pharmacology identified EHW's core bioactive components (quercetin, kaempferol, and β-sitosterol) and critical targets (IL-6, STAT3, HIF-1α, PTGS2, NFE2L2, and KEAP1) which were linked to ferroptosis and oxidative stress. Molecular docking revealed robust binding affinities between these compounds and ferroptosis-related proteins. In vivo validation confirmed that EHW inhibited KEAP1, activated NFE2L2-mediated antioxidant defenses (upregulating SOD1 and NQO1), restored iron homeostasis (lowering TFR1, elevating FTH1), and attenuated phospholipid peroxidation by suppressing ACSL4 and ALOX12. These results indicate that EHW mitigates ferroptosis-driven liver injury via KEAP1-NFE2L2 signaling to restore iron homeostasis and reduce oxidative stress, offering a mechanistic foundation for its clinical application in veterinary hepatoprotection.
Collapse
Affiliation(s)
- Hongxu Du
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
- Institute of Traditional Chinese Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Kunzhao Yang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Jingyi Yang
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Junjie Wan
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Yu Pan
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Weijie Song
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Shuang Xu
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Cheng Chen
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| | - Jiahui Li
- Department of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Southwest University, Chongqing 402460, China
| |
Collapse
|
50
|
Mishima E, Nakamura T, Doll S, Proneth B, Fedorova M, Pratt DA, Friedmann Angeli JP, Dixon SJ, Wahida A, Conrad M. Recommendations for robust and reproducible research on ferroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00843-2. [PMID: 40204928 DOI: 10.1038/s41580-025-00843-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
Ferroptosis is a necrotic, non-apoptotic cell death modality triggered by unrestrained iron-dependent lipid peroxidation. By unveiling the regulatory mechanisms of ferroptosis and its relevance to various diseases, research over the past decade has positioned ferroptosis as a promising therapeutic target. The rapid growth of this research field presents challenges, associated with potentially inadequate experimental approaches that may lead to misinterpretations in the assessment of ferroptosis. Typical examples include assessing whether an observed phenotype is indeed linked to ferroptosis, and selecting appropriate animal models and small-molecule modulators of ferroptotic cell death. This Expert Recommendation outlines state-of-the-art methods and tools to reliably study ferroptosis and increase the reproducibility and robustness of experimental results. We present highly validated compounds and animal models, and discuss their advantages and limitations. Furthermore, we provide an overview of the regulatory mechanisms and the best-studied players in ferroptosis regulation, such as GPX4, FSP1, SLC7A11 and ACSL4, discussing frequent pitfalls in experimental design and relevant guidance. These recommendations are intended for researchers at all levels, including those entering the expanding and exciting field of ferroptosis research.
Collapse
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Sebastian Doll
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany.
- Translational Redox Biology, TUM Natural School of Sciences, Technical University of Munich, Garching, Germany.
| |
Collapse
|