1
|
Gao YF, Yang YJ, Qin JB, Yu MY, Hu SW, Zhang HF, Lin FH, Hu HY, Fang MJ, Zeng JZ. Design, synthesis, and biological evaluation of quinolinyl-ureido-phenyl-hydrazide derivatives and quinolinyl-hydrazide derivatives as anticancer agents targeting Nur77-mediated ferroptosis. Eur J Med Chem 2025; 291:117559. [PMID: 40215561 DOI: 10.1016/j.ejmech.2025.117559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 05/03/2025]
Abstract
In the recent decade, targeting ferroptosis for cancer therapy has attracted remarkable attention. Interestingly, the transcriptional regulator Nur77, a promising therapeutic target in cancer, has been recently identified as a crucial regulator of ferroptosis. However, no ferroptosis inducer targeting Nur77 has been reported currently. In this study, we built upon our prior research on Nur77 modulator 4-PQBH to design and synthesize four series of new compounds, with the objective of developing novel Nur77-mediated ferroptosis inducers. Among them, compound 8f exhibited the most potency against the tested cancer cell lines, including human estrogen positive breast cancer and triple-negative breast cancer cell lines, while displaying lower toxicity towards human normal cell lines HaCaT and MCF-10A (IC50> 50 μM). Furthermore, 8f demonstrated superior Nur77-binding activity in comparison to the reference compound Csn-B, and it has the capacity to activate the Nur77-driven luciferase activity and increase the protein level of Nur77. Remarkably, 8f induced an increase in the levels of reactive oxygen species (ROS), malondialdehyde (MDA), and lipid peroxidation, concurrently with a reduction in the expression of GPX4 protein, culminating in the induction of ferroptosis in a Nur77-dependent manner. In vivo, 8f treatment has been observed to significantly suppress MCF7 xenograft tumor growth. Consequently, a novel ferroptosis inducer targeting Nur77 (8f) is first reported as a potent anti-EPBC agent, providing may serve as a promising lead for further drug development targeting Nur77-mediated ferroptosis.
Collapse
Affiliation(s)
- Yan-Fang Gao
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Yi-Jing Yang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jing-Bo Qin
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China; Guangdong Provincial Engineering Research Center of Molecular Imaging, Guangdong-Hong Kong-Macao University Joint Laboratory of the Interventional Medicine Foundation of Guangdong Province, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Ming-Yue Yu
- Xingzhi College, Zhejiang Normal University, Lanxi, 321004, China; College of Chemistry and Bioengineering, Yichun, 336000, China
| | - Sheng-Wei Hu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Hao-Fan Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Fan-Hong Lin
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Hong-Yu Hu
- Xingzhi College, Zhejiang Normal University, Lanxi, 321004, China.
| | - Mei-Juan Fang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China.
| | - Jin-Zhang Zeng
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
2
|
Chang Y, Li S, Chen K, Wang Y, Huang D, Wang X, Li J. Mitochondrial ferritin inhibition aggravates pacing-induced ventricular arrhythmias after myocardial infarction by promoting cardiomyocyte ferroptosis. Cell Signal 2025; 131:111683. [PMID: 40023300 DOI: 10.1016/j.cellsig.2025.111683] [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: 12/24/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Post-myocardial infarction ventricular arrhythmias are a leading cause of sudden cardiac death (SCD) following acute myocardial infarction worldwide. Emerging evidence suggests that ferroptosis, an iron-dependent form of cell death, plays a significant role in myocardial infarction damage. While mitochondrial ferritin (FtMt) is known to encapsulate harmful ferrous ions within mitochondria, its role in the development of post-myocardial infarction ventricular arrhythmias (post-MI VAs) is not well understood. OBJECTIVE This study aimed to clarify the role and mechanisms by which FtMt-mediated ferroptosis influences susceptibility to post-MI VAs. METHODS Mice were subjected to permanent ligation of the left anterior descending artery (LAD) to induce myocardial infarction (MI), followed by intracardiac electrophysiological studies to evaluate their vulnerability to post-MI VAs. Patch-clamp recordings and confocal Ca2+ imaging provided data on neonatal rat ventricular myocytes (NRVMs). We utilized DCFH-DA staining, transmission electron microscopy, and Seahorse analysis to examine the mitochondrial bioenergetics and oxidative phosphorylation in NRVMs. RESULTS Ferroptosis was activated in mice post-MI. Inhibiting ferroptosis enhanced cardiac function and reduced the incidence of post-MI VAs. Hypoxia led to electrophysiological dysregulation in NRVMs, which was exacerbated by FtMt inhibition. Specifically, FtMt inhibition under hypoxic conditions further impaired mitochondrial bioenergetics and oxidative phosphorylation, promoting ferroptosis in NRVMs. CONCLUSION FtMt plays a crucial protective function in MI by limiting infarct size, decreasing the frequency of ventricular arrhythmias, and inhibiting ferroptosis both in vivo and in vitro. These results suggest that FtMt may be a viable therapeutic target for treating MI.
Collapse
Affiliation(s)
- Yuchen Chang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, 200233 Shanghai, China
| | - Shuai Li
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, 200233 Shanghai, China
| | - Kankai Chen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, 200233 Shanghai, China
| | - Yanpeng Wang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, 200233 Shanghai, China
| | - Dong Huang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, 200233 Shanghai, China
| | - Xiaoqing Wang
- Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Jingbo Li
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, 200233 Shanghai, China.
| |
Collapse
|
3
|
Zhang J, Cai R, Ning C, Zhou Z, Zhang Y, Wang S, Li Y, Guo X. A novel integrated diagnostic and therapeutic ferroptosis inhibitor based on a phenothiazine scaffold with ROS-Responsive strategy. Bioorg Med Chem 2025; 124:118195. [PMID: 40252566 DOI: 10.1016/j.bmc.2025.118195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
Ferroptosis is a newly discovered form of cell death that is closely related to the occurrence of various diseases, such as neurodegenerative diseases, cardiovascular and cerebrovascular ischemic damage, and organ fibrosis. Therefore, the discovery of new active compounds with ferroptosis inhibitory activity is regarded as a new strategy for the clinical treatment of these diseases. In this study, a multifunctional prodrug molecule PNX-B2 with a phenoxazine structure was designed based on the oxidative microenvironment characteristic of ferroptosis. PNX-B2 can recognize the ferroptosis-associated oxidative conditions and simultaneously release compounds with ferroptosis-inhibitory activity. Moreover, it integrates diagnostic and therapeutic functions and offers a fluorescent indication of the ferroptosis microenvironment. PNX-B2 has demonstrated excellent ferroptosis-inhibitory activity with an EC50 value of 1.7 nM. This intelligent multifunctional compound shows great potential as a novel clinical agent for ferroptosis inhibition and presents broad prospects for future development.
Collapse
Affiliation(s)
- Jiangye Zhang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Rui Cai
- Instrumental Analysis Center, Dalian University of Technology, Dalian 116024, China.
| | - Changxu Ning
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Zhongxiang Zhou
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yibo Zhang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shisheng Wang
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Yueqing Li
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Xiuhan Guo
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; Ningbo Institute of Dalian University of Technology, Ningbo 315016, China.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Xie P, Qu T, Tang K, Huang Y, Zeng G, Yuan H, Xin Q, Zhao Y, Yang J, Zeng C, Wu X, Yang ST, Tang X. Carbon nanoparticles-Fe(II) complex combined with sorafenib for ferroptosis-induced antitumor effects in triple-negative breast cancer. Colloids Surf B Biointerfaces 2025; 250:114562. [PMID: 39965484 DOI: 10.1016/j.colsurfb.2025.114562] [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: 11/29/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Triple negative breast cancer (TNBC) represents an aggressive subtype of breast cancer that lacks the expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2, whose systemic treatment options are currently limited to chemotherapy. Carbon nanoparticles-Fe(II) complex (CNSI-Fe) is a promising antitumor drug that induces ferroptosis to kill tumor cells efficiently. In this study, we combined CNSI-Fe and a ferroptosis inducer sorafenib (SRF) to achieve the efficient chemotherapy of TNBC. CNSI-Fe could adsorb SRF by hydrophobic interaction and π-π stacking with a maximum adsorption capacity of 31 mg/g. During the in vitro assays, CNSI-Fe+SRF combination inhibited the cell viability of 4T1 cells much more efficiently than CNSI-Fe or SRF alone. The high Fe uptake, hydroxyl radical generation and oxidative damages verified the ferroptosis of 4T1 cells upon the CNSI-Fe+SRF treatment. During the in vivo evaluations, SRF enhanced the therapeutic effect of CNSI-Fe as indicated by the higher tumor growth inhibition rate of 67.8 % and the higher survival rate. CNSI captured SRF in tumor to give a 6 mg/kg uptake, which lowered the glutathione peroxidase 4 (GPX4) level and enhanced the hydroxyl radical production of 4T1 tumor. In addition, CNSI-Fe treatment up-regulated the genes associated with antioxidative responses, but the up-regulation was offset by SRF. CNSI-Fe+SRF group showed similar toxicity to mice as SRF alone in the biosafety evaluations. Our results collectively indicated that the combination of CNSI-Fe and SRF could efficiently treat TNBC through ferroptosis.
Collapse
Affiliation(s)
- Ping Xie
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China
| | - Ting Qu
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China
| | - Kexin Tang
- School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, China
| | - Yuanfang Huang
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China
| | - Guangfu Zeng
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China
| | - Huahui Yuan
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China
| | - Qian Xin
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China
| | - Yufeng Zhao
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China
| | - Jinmei Yang
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China
| | - Cheng Zeng
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China
| | - Xian Wu
- School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, China
| | - Sheng-Tao Yang
- School of Chemistry and Environment, Southwest Minzu University, Chengdu 610041, China.
| | - Xiaohai Tang
- Sichuan Enray Pharmaceutical Sciences Company, Chengdu 610095, China.
| |
Collapse
|
7
|
El-Naggar AM, Li Y, Turgu B, Ding Y, Wei L, Chen SY, Trigo-Gonzalez G, Kalantari F, Vallejos R, Lynch B, Senz J, Lum A, Douglas JM, Salamanca C, Thornton S, Qin Y, Parmar K, Spencer SE, Leung S, Woo MM, Yong PJ, Zhang HF, Hughes CS, Negri GL, Wang Y, Morin GB, Sorensen PH, Huntsman DG. Cystathionine gamma-lyase-mediated hypoxia inducible factor 1-alpha expression drives clear cell ovarian cancer progression. J Pathol 2025. [PMID: 40371821 DOI: 10.1002/path.6433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/07/2025] [Accepted: 03/26/2025] [Indexed: 05/16/2025]
Abstract
Clear cell ovarian cancer (CCOC) is the second most common ovarian cancer subtype, accounting for 5%-11% of ovarian cancers in North America. Late-stage CCOC is associated with a worse prognosis compared to other ovarian cancer histotypes, a challenge that has seen limited progress in recent decades. CCOC typically originates within the toxic microenvironment of endometriotic ovarian cysts and is characterized by its intrinsic chemoresistance, a strong hypoxic signature, and abundant expression of cystathionine gamma-lyase (CTH). CTH is a key enzyme in the transsulfuration pathway and serves as a marker of ciliated cells derived from the Müllerian tract. CTH plays a pivotal role in de novo cysteine synthesis, which is essential for glutathione (GSH) production and redox homeostasis. Using an array of molecular tools and cancer models, including in vivo studies, we demonstrated that CTH expression was induced under various stress conditions, such as exposure to endometriotic cyst content and hypoxia. This induction enables cell survival and creates a differentiation state manifested by CCOC that potentiates tumor progression and metastasis. In addition to regulating redox homeostasis, CTH enhances hypoxia inducible factor 1-alpha (HIF1α) expression, independently of hydrogen sulfide (H2S) production. Re-expression of HIF1α in CTH KO cells fully restored metastatic capacity in in vivo models. Co-expression of CTH and HIF1α proteins was also observed in human CCOC samples. Importantly, targeting CTH in CCOC significantly reduced its metastatic potential in in vivo models and enhanced sensitivity to chemotherapy. These findings underscore that CTH is both a defining feature of CCOC and a promising therapeutic target, not only for CCOC patients but also for those with other CTH-expressing cancers. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Amal M El-Naggar
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology, Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
| | - Yuqin Li
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Busra Turgu
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yuchen Ding
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Longyijie Wei
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Shary Yuting Chen
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Genny Trigo-Gonzalez
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Forouh Kalantari
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Rodrigo Vallejos
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Branden Lynch
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Janine Senz
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Amy Lum
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - J Maxwell Douglas
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Clara Salamanca
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Shelby Thornton
- Molecular and Advanced Pathology Core (MAPCore), University of British Columbia, Vancouver, BC, Canada
| | - Yimei Qin
- Molecular and Advanced Pathology Core (MAPCore), University of British Columbia, Vancouver, BC, Canada
| | - Kiran Parmar
- BC Centre for Pelvic Pain and Endometriosis, BC Women's Hospital, Vancouver, BC, Canada
| | - Sandra E Spencer
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Samuel Leung
- Genetic Pathology Evaluation Centre, Vancouver, BC, Canada
| | - Michelle Mm Woo
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
- BC's Gynecologic Cancer Research Program, OVCARE, Vancouver, BC, Canada
| | - Paul J Yong
- BC Centre for Pelvic Pain and Endometriosis, BC Women's Hospital, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Hai-Feng Zhang
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | | - Gian Luca Negri
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Yemin Wang
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gregg B Morin
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Poul H Sorensen
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - David G Huntsman
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Molecular and Advanced Pathology Core (MAPCore), University of British Columbia, Vancouver, BC, Canada
- Genetic Pathology Evaluation Centre, Vancouver, BC, Canada
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
- BC's Gynecologic Cancer Research Program, OVCARE, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
8
|
Jwa NS, Hwang BK. Ferroptosis in plant immunity. PLANT COMMUNICATIONS 2025; 6:101299. [PMID: 40057824 DOI: 10.1016/j.xplc.2025.101299] [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: 11/10/2024] [Revised: 01/15/2025] [Accepted: 03/05/2025] [Indexed: 04/13/2025]
Abstract
Plant cell death is mediated by calcium, iron, and reactive oxygen species (ROS) signaling in plant immunity. The reconstruction of a nucleotide-binding leucine-rich-repeat receptor (NLR) supramolecular structure, called the resistosome, is intimately involved in the hypersensitive response (HR), a type of cell death involved in effector-triggered immunity (ETI). Iron is a crucial redox catalyst in various cellular reactions. Ferroptosis is a regulated, non-apoptotic form of iron- and ROS-dependent cell death in plants. Pathogen infections trigger iron accumulation and ROS bursts in plant cells, leading to lipid peroxidation via the Fenton reaction and subsequent ferroptosis in plant cells similar to that in mammalian cells. The small-molecule inducer erastin triggers iron-dependent lipid ROS accumulation and glutathione depletion, leading to HR cell death in plant immunity. Calcium (Ca2+) is another major mediator of plant immunity. Cytoplasmic Ca2+ influx through calcium-permeable channels, the resistosomes, mediates iron- and ROS-dependent ferroptotic cell death under reduced glutathione reductase (GR) expression levels in the ETI response. Acibenzolar-S-methyl (ASM), a plant defense activator, enhances Ca2+ influx, ROS and iron accumulation, and lipid peroxidation to trigger ferroptotic cell death. These breakthroughs suggest a potential role for Ca2+ signaling in ferroptosis and its coordination with iron and ROS signaling in plant immunity. In this review, we highlight the essential roles of calcium, iron, and ROS signaling in ferroptosis during plant immunity and discuss advances in the understanding of how Ca2+-mediated ferroptotic cell death orchestrates effective plant immune responses against invading pathogens.
Collapse
Affiliation(s)
- Nam-Soo Jwa
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul 05006, Korea.
| | - Byung Kook Hwang
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea.
| |
Collapse
|
9
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Mechanism of ferroptosis in heart failure: The role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and intervention strategies. Ageing Res Rev 2025:102770. [PMID: 40360081 DOI: 10.1016/j.arr.2025.102770] [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: 12/03/2024] [Revised: 04/29/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
The ferroptosis of cardiomyocytes has been recognized as the core pathological mechanism of heart failure. During the evolution of cardiovascular diseases, the accumulation of angiotensin II and advanced glycation end products can lead to the excessive activation of the RAGE/TLR4-JNK1/2 pathway, which subsequently triggers ferritinophagy, clockophagy, and enhanced p53 activity, ultimately leading to cardiomyocyte ferroptosis. It is evident that deeply unraveling the specific mechanisms in this field and comprehensively evaluating potential drugs and therapeutic strategies targeting this pathway is crucial for improving the status of cardiomyocyte ferroptosis. However, our current understanding of this pathway's specific molecular biological mechanisms in the process of cardiomyocyte ferroptosis remains limited. In light of this, this paper first comprehensively reviews the historical context of ferroptosis research, compares the similarities and differences between ferroptosis and other standard modes of cell death, elucidates the core mechanisms of ferroptosis and its close connection with heart failure, aiming to establish a basic cognitive framework for readers on ferroptosis and its role in heart failure. Subsequently, the paper delves into the pivotal role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and its intricate molecular biological regulatory network. Furthermore, it systematically integrates various therapeutic approaches aimed at inhibiting RAGE, TLR4, and JNK1/2 activity to alleviate cardiomyocyte ferroptosis, encompassing RNA interference technology, gene knockout techniques, small molecule inhibitors, natural active ingredients, as well as traditional Chinese and Western medicines, with the ultimate goal of forging new avenues and strategies for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| |
Collapse
|
10
|
Su Y, Liu X, Wang D, Li G, Mi X, Zhang Y, Yue S, Zhang Z, Shen T, Tan X. Salinomycin inhibits SREBP1 to sensitize ferroptosis and ameliorate sorafenib resistance in clear cell renal cell carcinoma. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025:119989. [PMID: 40360020 DOI: 10.1016/j.bbamcr.2025.119989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 04/20/2025] [Accepted: 05/09/2025] [Indexed: 05/15/2025]
Abstract
Backgrounds Resistance to sorafenib, a frontline therapy for advanced ccRCC, is associated with decreased sensitivity to ferroptosis. Our research focuses on elucidating the mechanisms underlying ccRCC's resistance to sorafenib-induced ferroptosis and identifying potential new agents that could overcome this resistance. METHODS The silencing of SREBP1 was employed to evaluate the role of this key transcription factor in lipid synthesis and its contribution to ferroptosis resistance in sorafenib-treated ccRCC cells. The ATF4-mediated induction of SREBP1 following Salinomycin treatment was assessed by western blot, RT-PCR, immunohistochemistry, chromatin immunoprecipitation, and dual-luciferase reporter assays. In cultured ccRCC cells, the combined effects of Salinomycin and sorafenib on ferroptosis induction were evaluated by assessing cell viability, glutathione levels, malondialdehyde levels, BODIPY fluorescence, and intracellular Fe2+ concentration. In an orthotopic ccRCC mouse model, the synergistic effects of Salinomycin and sorafenib on both ferroptosis and tumor progression were examined. RESULTS Overexpression of SREBP1 was observed in ccRCC tumor tissue, and induced by sorafenib treatment. Silencing SREBP1 reduced the resistance of ccRCC cells to ferroptosis induced by sorafenib. Salinomycin decreased ATF4 level, which in turn inhibited SREBP1 transcription. Treatment with salinomycin enhanced the sensitivity of ccRCC cells to sorafenib-induced ferroptosis. In the orthotopic xenograft mouse model of ccRCC, the combination of salinomycin and sorafenib showed a synergistic effect in inducing ferroptosis inhibiting tumor growth. CONCLUSIONS Salinomycin treatment mitigates resistance to sorafenib-induced ferroptosis by inhibiting SREBP1. The combination of salinomycin and sorafenib synergistically enhances ferroptosis and suppresses ccRCC growth.
Collapse
Affiliation(s)
- Yu Su
- The School of Medicine, Nankai University; 94 Wei Jin Road, Tianjin, China
| | - Xuan Liu
- The School of Medicine, Nankai University; 94 Wei Jin Road, Tianjin, China
| | - Dekun Wang
- The School of Medicine, Nankai University; 94 Wei Jin Road, Tianjin, China
| | - Gang Li
- Department of Urology, Tianjin Institute of Urology, the 2nd Hospital of Tianjin Medical University, 23 Pingjiang Road, Tianjin, China
| | - Xue Mi
- The School of Medicine, Nankai University; 94 Wei Jin Road, Tianjin, China
| | - Yuying Zhang
- The School of Medicine, Nankai University; 94 Wei Jin Road, Tianjin, China
| | - Shijing Yue
- The School of Medicine, Nankai University; 94 Wei Jin Road, Tianjin, China
| | - Zhujun Zhang
- The School of Medicine, Nankai University; 94 Wei Jin Road, Tianjin, China
| | - Tianyu Shen
- The School of Medicine, Nankai University; 94 Wei Jin Road, Tianjin, China.
| | - Xiaoyue Tan
- The School of Medicine, Nankai University; 94 Wei Jin Road, Tianjin, China.
| |
Collapse
|
11
|
Cui L, Xia Q, Wang Y, Han C, Zang X, Zhang L, Xing J, Zheng R, Zhang Y. Luteolin-7-O-glucuronide alleviates doxorubicin-induced cardiotoxicity by inhibiting PPAR-mediated ferroptosis. Toxicol Appl Pharmacol 2025; 500:117381. [PMID: 40345555 DOI: 10.1016/j.taap.2025.117381] [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: 01/26/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Inhibiting ferroptosis has been proposed to rescue myocardial cell death in Doxorubicin (DOX)-induced cardiotoxicity (DIC). Here, we aimed to investigate whether luteolin-7-O-glucuronide (LOG) alleviates DIC via ferroptosis suppression in zebrafish and H9C2 cardiomyocytes, as well as the potential mechanism. We found that LOG improved zebrafish cardiac function and mitigated the upregulation of CK-MB, cTnT, nppa, and nppb caused by DOX. Moreover, LOG suppressed the high levels of ROS, GSSG, and MDA in response to DOX and increased GSH activity and gpx4 levels in zebrafish. Additionally, LOG increased cell viability and the GSH/GSSG ratio, reduced oxidative damage and the accumulation of ferrous ions, and maintained mitochondrial function in H9C2 cells. Mechanistically, LOG improved the abnormal expression of key genes in the PPAR signaling pathway and ferroptosis induced by DOX. In conclusion, our study emphasized that LOG attenuates DIC by mitigating oxidative stress-triggered lipid peroxidation related to the inhibition of PPAR-mediated ferroptosis.
Collapse
Affiliation(s)
- Lishuang Cui
- Xinjiang Medical University, Urumqi 830054, China
| | - Qing Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Yongcheng Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Cong Han
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Xiaohan Zang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China
| | - Lijuan Zhang
- Xinjiang Key Laboratory of Uygur Medical Research, Xinjiang Institute of Materia Medica, Urumqi 841100, China
| | - Jianguo Xing
- Xinjiang Medical University, Urumqi 830054, China; Xinjiang Key Laboratory of Uygur Medical Research, Xinjiang Institute of Materia Medica, Urumqi 841100, China.
| | - Ruifang Zheng
- Xinjiang Key Laboratory of Uygur Medical Research, Xinjiang Institute of Materia Medica, Urumqi 841100, China; Hetian Uygur Pharmaceutical Limited Liablity Company, Hetian 848200, China.
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China.
| |
Collapse
|
12
|
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
|
13
|
An Y, Liu W, Deng Y, Huang W, Huang J. SLC7A11-HSPB1 Axis: A Novel Mechanism for Hepatocellular Carcinoma Progression and Ferroptosis Regulation. Biomed J 2025:100869. [PMID: 40339903 DOI: 10.1016/j.bj.2025.100869] [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: 11/19/2024] [Revised: 03/27/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND SLC7A11, a plasma membrane protein, has been implicated as an oncogene in various cancers, including hepatocellular carcinoma (HCC). Its role in HCC pathogenesis, particularly in relation to ferroptosis, is not well understood. This study aims to investigate the function of SLC7A11 with ferroptosis and its interaction in development of HCC. METHODS AND MATERIAL Clinical HCC tissue samples were used to analyze the expression of SLC7A11 by RT-PCR. The impact of SLC7A11 on HCC cell viability, proliferation, and migration was assessed by CCK-8, AlamarBlue and Transwell. Protein-protein interactions were explored using co-immunoprecipitation and immunofluorescence. The effect of SLC7A11 on ferroptosis was evaluated by iron levels, ROS, and GSH. The impact of sorafenib and doxorubicin (DOX) on HCC cells was analyzed using cell viability assay. RESULTS SLC7A11 was found to be highly expressed in HCC tissues and was correlated with tumor size and poor prognosis. Overexpression of SLC7A11 in HCC cells promoted cell viability, proliferation, and migration. Additionally, SLC7A11 overexpression mitigated erastin-induced ferroptosis, as evidenced by decreased ROS levels and increased GSH levels. We also discovered that SLC7A11 interacted with HSPB1. HSPB1 inhibited erastin-induced ferroptosis. Furthermore, a portion of the cell death induced by sorafenib and DOX is attributed to ferroptosis, with HSPB1 and SLC7A11 inhibiting the death induced by the two drugs, respectively. CONCLUSIONS SLC7A11 plays a significant role in HCC progression by inhibiting ferroptosis, and its interaction with HSPB1 is a critical pathway in this process. Targeting the SLC7A11-HSPB1 axis may provide a novel therapeutic strategy for HCC treatment, highlighting the importance of understanding the mechanisms of ferroptosis in cancer cells.
Collapse
Affiliation(s)
- Yan An
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China; Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200050, China
| | - Weilong Liu
- Institute of Hepatology, National clinical research center for infectious diseases, Guangdong Key Lab for Diagnosis &Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
14
|
Ju Y, Zhang Y, Tian X, Zhu N, Zheng Y, Qiao Y, Yang T, Niu B, Li X, Yu L, Liu Z, Wu Y, Zhi Y, Dong Y, Xu Q, Yang X, Wang X, Wang X, Deng H, Mao Y, Li X. Protein S-glutathionylation confers cellular resistance to ferroptosis induced by glutathione depletion. Redox Biol 2025; 83:103660. [PMID: 40354766 DOI: 10.1016/j.redox.2025.103660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 04/29/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025] Open
Abstract
Ferroptosis is one of the most critical biological consequences of glutathione depletion. Excessive oxidative stress, indicated by an elevated oxidized glutathione (GSSG)/reduced glutathione (GSH) ratio, is recognized as a key driver of ferroptosis. However, in glutathione depletion-induced ferroptosis, a marked decrease in total glutathione levels (including both GSH and GSSG) is frequently observed, yet its significance remains understudied. Protein S-glutathionylation (protein-SSG) levels are closely linked to the redox state and cellular glutathione pools including GSH and GSSG. To date, the role of protein-SSG during cell ferroptosis induced by glutathione depletion remains poorly understood. Here, we demonstrated that upregulation of CHAC1, a glutathione-degrading enzyme, acted as a key regulator of protein-SSG formation and exacerbated glutathione depletion-induced ferroptosis. This effect was observed in both in vitro and in vivo models, including erastin-induced ferroptosis across multiple cell lines and acetaminophen overdose-triggered ferroptosis in hepatocytes. Deficiency of CHAC1 resulted in increased glutathione pools, enhanced protein-SSG, improved liver function, and attenuation of hepatocyte ferroptosis upon acetaminophen challenge. These protective effects were reversed by CHAC1 overexpression. Using quantitative redox proteomics, we identified glutathione pool-sensitive S-glutathionylated proteins. As an important example, we discovered that ADP-ribosylation factor 6 (ARF6) was regulated by S-glutathionylation during glutathione depletion-induced ferroptosis. Our findings revealed that CHAC1 upregulation reduced the S-glutathionylation of ARF6, resulting in decreased ARF6 levels in lysosomes. This, in turn, enhanced the localization of the transferrin receptor (TFRC) on the cell membrane and increased transferrin uptake, ultimately compromising the protective role of ARF6 in ferroptosis induced by glutathione depletion. Targeting TFRC using GalNAc-siTfrc mitigated acetaminophen-induced liver injury in vivo. In conclusion, our study provide evidence that availability of glutathione pools affects protein S-glutathionylation and regulates protein functions to influence the process of ferroptosis, which opens an avenue to understanding the cell ferroptosis induced by glutathione depletion.
Collapse
Affiliation(s)
- Yi Ju
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuting Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaolin Tian
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nanbin Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yufan Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiming Qiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tao Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Baolin Niu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaoyun Li
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, NHC Key Laboratory of Digestive Diseases, Shanghai Research Center of Fatty Liver Disease, Shanghai, China
| | - Liu Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhuolin Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yixuan Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yang Zhi
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, NHC Key Laboratory of Digestive Diseases, Shanghai Research Center of Fatty Liver Disease, Shanghai, China
| | - Yinuo Dong
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, NHC Key Laboratory of Digestive Diseases, Shanghai Research Center of Fatty Liver Disease, Shanghai, China
| | - Qingling Xu
- Department of Hepatology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaoming Yang
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xuening Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xiaokai Wang
- Department of Vascular Surgery, Xuzhou First People's Hospital, Xuzhou, Jiangsu, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yimin Mao
- Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, NHC Key Laboratory of Digestive Diseases, Shanghai Research Center of Fatty Liver Disease, Shanghai, China.
| | - Xiaobo Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Liu K, Wang Z, Guo X, Luo J, Wu X, Wang F, Mei Y. The glutamine starvation-induced lncRNA FERRIN suppresses ferroptosis via the stabilization of SLC7A11 mRNA. Int J Biol Macromol 2025; 308:142388. [PMID: 40127798 DOI: 10.1016/j.ijbiomac.2025.142388] [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: 09/20/2024] [Revised: 11/19/2024] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
As an essential nutrient for cancer cell survival, glutamine plays both promoting and inhibitory roles in ferroptosis; however, the underlying mechanisms remain obscure. Emerging evidence suggests that long noncoding RNAs (lncRNAs) are crucial regulators of ferroptosis. Nevertheless, it remains unclear whether lncRNAs are involved in glutamine-regulated ferroptosis. In this study, we report that the lncRNA FERRIN is induced by the transcription factor ATF4 under glutamine starvation conditions. FERRIN functions as an inhibitor of ferroptosis by upregulating SLC7A11 expression. Mechanistically, FERRIN interacts with the RNA binding protein hnRNPK, facilitating its binding to SLC7A11 mRNA and leading to the stabilization of SLC7A11 mRNA. Consistent with its inhibitory role in ferroptosis, FERRIN promotes in vitro cancer cell proliferation and in vivo xenograft tumor growth through its modulation of SLC7A11. Collectively, these findings establish FERRIN as a critical negative regulator of ferroptosis and suggest that FERRIN may represent an important link between glutamine availability and ferroptosis.
Collapse
Affiliation(s)
- Kaiyue Liu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhongyu Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaorui Guo
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jingjing Luo
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xianning Wu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Fang Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.
| | - Yide Mei
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Wood JPM, Chidlow G, Casson RJ. Glucose protects cultured retinal cells from oxidative injury via the pentose phosphate pathway. Free Radic Biol Med 2025; 232:142-157. [PMID: 40054635 DOI: 10.1016/j.freeradbiomed.2025.03.006] [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: 09/25/2024] [Revised: 02/16/2025] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
PURPOSE Oxidative injury has been implicated in a range of common retinal neurodegenerative disorders. Protecting the retina from such an insult could therefore prove clinically beneficial. We sought to investigate whether glucose, acting via the pentose phosphate pathway (PPP), was able to counteract oxidative cytotoxicity to retinal cells in culture. EXPERIMENTAL Mixed retinal neuron-glial cultures were prepared from Sprague-Dawley rat neonates and used at 7 days in vitro; neuron-only and Müller glial cell-only mono-cultures were subsequently prepared from these cultures. At appropriate stages, cultures were treated with t-butyl hydroperoxide (tbH; 10 nM-1 mM) in glucose/pyruvate-free DMEM to induce oxidative stress. Some cultures were co-treated with glucose. Additional compounds were co-applied to inhibit glycolysis, PPP, cystine uptake, glutathione biosynthesis and glutathione reductase (GR). The effect of glucose on stimulation of reactive oxygen species (ROS), as well as levels of glutathione and NADPH were also investigated. RESULTS Oxidative stress resulted in cytotoxicity to both retinal neurons and glial cells. Glucose was able to abrogate the toxicity to glial cells in mono-cultures and mixed cultures, but could only provide protection to neurons in the mixed cultures when glial cells were also present. Glucose was additionally shown to prevent stimulation of ROS and oxidative stress-induced depletions of glutathione and NADPH. Inhibition of PPP, cystine uptake or GR all diminished the protective response of glucose. CONCLUSION Glucose prevented oxidative stress to retinal cells via the PPP. Neurons were not subjected to glucose-induced protection except when glial cells were present, implying the passage of a transmissible mediator or other protective action between the two cell types.
Collapse
Affiliation(s)
- John P M Wood
- Discipline of Ophthalmology & Visual Sciences, Level 7 Adelaide Health and Medical Sciences Building, University of Adelaide, North Terrace, Adelaide, SA, 5000, Australia; South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Port Road, SA, 5000, Australia.
| | - Glyn Chidlow
- Discipline of Ophthalmology & Visual Sciences, Level 7 Adelaide Health and Medical Sciences Building, University of Adelaide, North Terrace, Adelaide, SA, 5000, Australia; South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Port Road, SA, 5000, Australia
| | - Robert J Casson
- Discipline of Ophthalmology & Visual Sciences, Level 7 Adelaide Health and Medical Sciences Building, University of Adelaide, North Terrace, Adelaide, SA, 5000, Australia; South Australian Institute of Ophthalmology, Royal Adelaide Hospital, Port Road, SA, 5000, Australia
| |
Collapse
|
19
|
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
|
20
|
Liu Y, Yao Y, Yu Y, Sun Y, Wu M, Chen R, Feng H, Guo S, Yang Y, Zhang C. Role of T 3 in the Regulation of GRP78 on Granulosa Cells in Rat Ovaries. Int J Mol Sci 2025; 26:4196. [PMID: 40362432 PMCID: PMC12072174 DOI: 10.3390/ijms26094196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 04/25/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Thyroid hormone (TH) plays a vital role in ovarian follicle development, and glucose-regulated protein 78 (GRP78) is involved in these processes, which is regulated by TH. However, the mechanisms are still unclear. To evaluate the possible mechanism of TH on the regulation of GRP78 expression, Cleavage Under Targets and Tagmentation (CUT & Tag) sequencing, luciferase assays, and Electrophoretic Mobility Shift Assays (EMSA) were employed to delineate the binding sites of thyroid hormone receptor β (TRβ) on the GRP78 promoter and to confirm the interactions. Additionally, Co-Immunoprecipitation (Co-IP) and Immunofluorescence (IF) assays were used to investigate the interactions between TRβ and the coactivator peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) after triiodothyronine (T3) treatment with different concentrations. Our findings identified a thyroid hormone response element (TRE) on the GRP78 promoter and demonstrated that TRβ can activate GRP78 expression by interacting with PGC-1α. In order to simulate the condition of hyperthyroidism, granulosa cells (GCs) extracted from rats were treated by T3 with high concentrations, which decreased the expression of PGC-1α, resulting in decreased expressions of GRP78 and other ferroptosis-related markers such as glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11, xCT), thereby inducing ferroptosis in GCs. Taken together, the present study demonstrates that T3 induces cellular ferroptosis by binding TRE of the GRP78 promoter in ovarian GCs via TRβ. As a switcher, PGC-1α is also involved in these processes.
Collapse
Affiliation(s)
- Yan Liu
- College of Life Science, Capital Normal University, Beijing 100048, China; (Y.L.); (Y.Y.); (Y.Y.); (Y.S.); (M.W.); (R.C.); (H.F.); (S.G.)
| | - Yilin Yao
- College of Life Science, Capital Normal University, Beijing 100048, China; (Y.L.); (Y.Y.); (Y.Y.); (Y.S.); (M.W.); (R.C.); (H.F.); (S.G.)
| | - Yakun Yu
- College of Life Science, Capital Normal University, Beijing 100048, China; (Y.L.); (Y.Y.); (Y.Y.); (Y.S.); (M.W.); (R.C.); (H.F.); (S.G.)
| | - Ying Sun
- College of Life Science, Capital Normal University, Beijing 100048, China; (Y.L.); (Y.Y.); (Y.Y.); (Y.S.); (M.W.); (R.C.); (H.F.); (S.G.)
| | - Mingqi Wu
- College of Life Science, Capital Normal University, Beijing 100048, China; (Y.L.); (Y.Y.); (Y.Y.); (Y.S.); (M.W.); (R.C.); (H.F.); (S.G.)
| | - Rui Chen
- College of Life Science, Capital Normal University, Beijing 100048, China; (Y.L.); (Y.Y.); (Y.Y.); (Y.S.); (M.W.); (R.C.); (H.F.); (S.G.)
| | - Haoyuan Feng
- College of Life Science, Capital Normal University, Beijing 100048, China; (Y.L.); (Y.Y.); (Y.Y.); (Y.S.); (M.W.); (R.C.); (H.F.); (S.G.)
| | - Shuaitian Guo
- College of Life Science, Capital Normal University, Beijing 100048, China; (Y.L.); (Y.Y.); (Y.Y.); (Y.S.); (M.W.); (R.C.); (H.F.); (S.G.)
| | - Yanzhou Yang
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, Ningxia Medical University, Yinchuan 750004, China
| | - Cheng Zhang
- College of Life Science, Capital Normal University, Beijing 100048, China; (Y.L.); (Y.Y.); (Y.Y.); (Y.S.); (M.W.); (R.C.); (H.F.); (S.G.)
| |
Collapse
|
21
|
Mavaddatiyan L, Naeini S, Khodabandeh S, Hosseini F, Skelton RP, Azizi V, Talkhabi M. Exploring the association between aging, ferroptosis, and common age-related diseases. Arch Gerontol Geriatr 2025; 135:105877. [PMID: 40339241 DOI: 10.1016/j.archger.2025.105877] [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: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/26/2025] [Indexed: 05/10/2025]
Abstract
Aging is a natural biological process that is characterized by the progressive decline in physiological functions and an increased vulnerability to age-related diseases. The aging process is driven by different cell and molecular mechanisms. It has recently been shown that aging is associated with heightened vulnerability to ferroptosis (an intracellular iron-dependent form of programmed cell death). This susceptibility arises from various factors including oxidative stress, impaired antioxidant defences, and dysregulated iron homeostasis. The progressive decline in cellular antioxidant capacity and the accumulation of damaged components contribute to the increased susceptibility of aging cells to ferroptosis. Dysregulation of key regulators involved in ferroptosis, such as glutathione peroxidase 4 (GPX4), iron regulatory proteins, and lipid metabolism enzymes, further exacerbates this vulnerability. The decline in cellular defence mechanisms against ferroptosis during aging contributes to the accumulation of damaged cells and tissues, ultimately resulting in the manifestation of age-related diseases. Understanding the intricate relevance between aging and ferroptosis holds significant potential for developing strategies to counteract the detrimental effects of aging and age-related diseases. This will subsequently act to mitigate the negative consequences of aging and improving overall health in the elderly population. This review aims to clarify the relationship between aging and ferroptosis, and explores the underlying mechanisms and implications for age-related disorders, including neurodegenerative, cardiovascular, and neoplastic diseases. We also discuss the accumulating evidence suggesting that the imbalance of redox homeostasis and perturbations in iron metabolism contribute to the age-associated vulnerability to ferroptosis.
Collapse
Affiliation(s)
- Laleh Mavaddatiyan
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - SaghiHakimi Naeini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Sara Khodabandeh
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Fatemeh Hosseini
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - RhysJ P Skelton
- Flinders Medical Centre, Department of Ophthalmology, Bedford Park, Australia
| | - Vahid Azizi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Mahmood Talkhabi
- Department of Animal Sciences and Marine Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
22
|
Paul B, Merta H, Ugrankar-Banerjee R, Hensley MR, Tran S, do Vale GD, Zacherias L, Hewett CK, McDonald JG, Font-Burgada J, Mathews TP, Farber SA, Henne WM. Paraoxonase-like APMAP maintains endoplasmic-reticulum-associated lipid and lipoprotein homeostasis. Dev Cell 2025:S1534-5807(25)00210-2. [PMID: 40318637 DOI: 10.1016/j.devcel.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 11/01/2024] [Accepted: 04/09/2025] [Indexed: 05/07/2025]
Abstract
Oxidative stress perturbs lipid homeostasis and contributes to metabolic diseases. Though ignored when compared with mitochondrial oxidation, the endoplasmic reticulum (ER) generates reactive oxygen species requiring antioxidant quality control. Using multi-organismal profiling featuring Drosophila, zebrafish, and mammalian hepatocytes, here we characterize the paraoxonase-like C20orf3/adipocyte plasma-membrane-associated protein (APMAP) as an ER-localized antioxidant that suppresses ER lipid oxidation to safeguard ER function. APMAP-depleted cells exhibit defective ER morphology, ER stress, and lipid peroxidation dependent on ER-oxidoreductase 1α (ERO1A), as well as sensitivity to ferroptosis and defects in ApoB-lipoprotein homeostasis. Similarly, organismal APMAP depletion in Drosophila and zebrafish perturbs ApoB-lipoprotein homeostasis. Strikingly, APMAP loss is rescued with chemical antioxidant N-acetyl-cysteine (NAC). Lipidomics identifies that APMAP loss elevates phospholipid peroxidation and boosts ceramides-signatures of lipid stress. Collectively, we propose that APMAP is an ER-localized antioxidant that promotes lipid and lipoprotein homeostasis in the ER network.
Collapse
Affiliation(s)
- Blessy Paul
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Holly Merta
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Monica R Hensley
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Son Tran
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Goncalo Dias do Vale
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lauren Zacherias
- Children's Research Institute (CRI), University of Texas Southwestern Medical, Dallas, TX 75390, USA
| | - Charles K Hewett
- Fox Chase Cancer Center, Temple Health, Philadelphia, PA 19111, USA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Thomas P Mathews
- Children's Research Institute (CRI), University of Texas Southwestern Medical, Dallas, TX 75390, USA
| | - Steven A Farber
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - W Mike Henne
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
23
|
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
|
24
|
He Z, Long H, Ma H, Ran W, Song Y, Shang W, Wang J, Qin Y, Deng Z. 4, 9-dihydroxy-α-lapachone as a potent antiproliferation agent for triple-negative breast cancer via ferroptosis. Fitoterapia 2025; 183:106567. [PMID: 40280251 DOI: 10.1016/j.fitote.2025.106567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant breast cancer. Ferroptosis is an oxidative, iron-dependent form of regulated cell death. Ferroptosis-targeted therapies is a promising approach to improving treatment outcomes of TNBC. Combining death pathway inhibitors with relevant indices for ferroptosis and LipROS, this study uncovered that a natural product of 4, 9-dihydroxy-α-lapachone (DLN) from Catalpa bungei "jinsi" exhibited in vitro and in vivo inhibitory activity against TNBC via ferroptosis. The molecular mechanism is an activation of the FTH1 led to iron overload, and then inhibition of cysteine-glutamate antiporter (system Xc-) and GPX4, which further sensitized TNBC cells to ferroptosis. This study clarified the pathway of DLN-induced cell death in TNBC treatment and exhibited its potential as therapeutic agent for TNBC.
Collapse
Affiliation(s)
- Zhong He
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hui Long
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Hongyue Ma
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Wanrong Ran
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yinhong Song
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, China.
| | - Wenjuan Shang
- Medical College, Hubei Three Gorges Polytechnic, Yichang 443000, China
| | - Junhui Wang
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation of State Forestry Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing 100091, China.
| | - Ye Qin
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Zhangshuang Deng
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
25
|
Kolligundla LP, Sullivan KM, Mukhi D, Andrade-Silva M, Liu H, Guan Y, Gu X, Wu J, Doke T, Hirohama D, Guarnieri P, Hill J, Pullen SS, Kuo J, Inamoto M, Susztak K. Glutathione-specific gamma-glutamylcyclotransferase 1 ( CHAC1) increases kidney disease risk by modulating ferroptosis. Sci Transl Med 2025; 17:eadn3079. [PMID: 40267214 DOI: 10.1126/scitranslmed.adn3079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 08/20/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Genome-wide association studies (GWASs) have identified more than 1000 loci where genetic variants correlate with kidney function. However, the specific genes, cell types, and mechanisms influenced by these genetic variants remain largely uncharted. Here, we identified glutathione-specific gamma-glutamylcyclotransferase 1 (CHAC1) on chromosome 15 as affected by GWAS variants by analyzing human kidney gene expression and methylation information. Both CHAC1 RNA and protein were expressed in the loop of Henle region in mouse and human kidneys, and CHAC1 expression was higher in patients carrying disease risk variants. Using CRISPR technology, we created mice with a single functional copy of the Chac1 gene (Chac1+/-) that displayed no baseline phenotypic alterations in kidney structure or function. These mice demonstrated resilience to kidney disease in multiple models, including folic acid-induced nephropathy, adenine-induced chronic kidney disease, and uninephrectomy-streptozotocin-induced diabetic nephropathy. We further showed that CHAC1 plays a critical role in degrading the cellular antioxidant glutathione. Tubule cells isolated from Chac1+/- mice showed increased glutathione, decreased lipid peroxidation, improved cell viability, and protection against ferroptosis. Expression of ferroptosis-associated genes was also lower in mice with only one copy of Chac1. Higher CHAC1 protein also correlated with ferroptosis-related protein abundance in kidney biopsies from patients with kidney disease. This study positions CHAC1 as an important mediator of kidney disease that influences glutathione concentrations and ferroptosis, suggesting potential avenues to explore for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Lakshmi P Kolligundla
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Katie M Sullivan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Pediatrics, Medical College of Wisconsin Pediatric Nephrology, Milwaukee, WI 53226, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Magaiver Andrade-Silva
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Hongbo Liu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Yuting Guan
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Xiangchen Gu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Junnan Wu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Daigoro Hirohama
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| | - Paolo Guarnieri
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jon Hill
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Steven S Pullen
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | - Jay Kuo
- Department of Cardiometabolic Diseases Research, Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT 06877, USA
| | | | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Penn/CHOP Kidney Innovation Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19014, USA
| |
Collapse
|
26
|
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
|
27
|
Sun H, Xu C, Xiong Z, Liu M, Ning X, Zhuang Y. Therapeutic prospects and potential mechanisms of Prdx6: as a novel target in musculoskeletal disorders. Front Physiol 2025; 16:1524100. [PMID: 40313876 PMCID: PMC12043587 DOI: 10.3389/fphys.2025.1524100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
With the global population aging, musculoskeletal disorders (MSDs) have posed significant physical and psychological health challenges for patients as well as a substantial economic burden on society. The advancements in conservative and surgical interventions for MSDs have been remarkable in recent years; however, the current treatment modalities still fall short of meeting the optimal requirements of patients. Recently, peroxiredoxin 6 (Prdx6) has gained considerable attention from researchers due to its remarkable antioxidative, anti-inflammatory, and anti-apoptotic properties. It has been found that Prdx6 is involved in multiple system diseases, including MSDs; however, the exact role of Prdx6 in MSDs is still lacking. This study aimed to summarize the structure, regulatory mechanism, and potential function of Prdx6. These findings may demonstrate Prdx6 as a novel target for inhibiting the advancement of MSDs.
Collapse
Affiliation(s)
- Hong Sun
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chao Xu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Zhilin Xiong
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Miao Liu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xu Ning
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yong Zhuang
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
28
|
Kaftan Öcal G, Armagan G. Induction of Ferroptotic Cell Death by Neuromelanin Pigments in Dopaminergic Cells. ACS Chem Neurosci 2025; 16:1500-1510. [PMID: 40145657 PMCID: PMC12006986 DOI: 10.1021/acschemneuro.5c00029] [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: 01/13/2025] [Revised: 02/24/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Neuromelanin (NM) is an iron-rich, insoluble brown or black pigment that exhibits protective properties. However, its accumulation over time may render it a source of free radicals. In Parkinson's disease, dopaminergic neurons with the highest NM levels and increased iron content are preferentially vulnerable to degeneration. Considering NM's iron binding capacity and the critical role of iron in ferroptosis, we aimed to investigate the interplay between neuromelanin and ferroptosis in dopaminergic cells. We prepared two NM pigments: iron-free NM (ifNM) and iron-containing NM (Fe3+NM) and, exposed to cells. After verifying NM accumulation, cell viability was assessed in the absence or presence of antioxidants (NAC (1 mM), Trolox (100 μM)) and specific inhibitors of cell death types. Ferroptosis-related parameters, including lipid peroxidation byproducts (4-HNE), lipid ROS, glutathione, intracellular iron, GPX4, and ACSL4, and cellular iron metabolism-related proteins (TfR1, ferroportin, ferritin, IREB2) were evaluated following ifNM and Fe3+NM treatments, with or without Ferrostatin-1, Liproxstatin-1 and deferoxamine. Both NMs induced cell death via distinct mechanisms. Ferroptotic cell death by ifNM and Fe3+NM was reversed by ferrostatin-1 and NAC (p < 0.05). Significant alterations in lipid peroxidation, GPX4 levels, and iron metabolism were observed independent of NM's iron composition (p < 0.05). Ferritin levels increased following ifNM treatment, reflecting an adaptive response to iron overload, while Fe3+NM treatment led to ferritin depletion, possibly via ferritinophagy. Our findings reveal a distinct role of iron-rich and iron-free neuromelanin in modulating ferroptotic pathways, highlighting the potential of targeting neuromelanin-iron interactions as a therapeutic strategy to mitigate neuronal ferroptosis in Parkinson's disease.
Collapse
Affiliation(s)
- Gizem Kaftan Öcal
- Biochemistry
PhD Programme, Graduate School of Health Sciences, Ege University, Izmir 35100, Türkiye
- Department
of Biochemistry, Faculty of Pharmacy, Ayfonkarahisar
Health Sciences University, Afyonkarahisar 03218, Türkiye
| | - Güliz Armagan
- Department
of Biochemistry, Faculty of Pharmacy, Ege
University, Izmir 35100, Türkiye
| |
Collapse
|
29
|
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
|
30
|
Wen B, Chang W, Yang L, Lv D, Wang L, Wang L, Xu Y, Hu J, Ding K, Xue Q, Qi X, Yang B, Wang J. The long noncoding RNA APR attenuates PPRV infection-induced accumulation of intracellular iron to inhibit membrane lipid peroxidation and viral replication. mBio 2025; 16:e0012725. [PMID: 40126010 PMCID: PMC11980570 DOI: 10.1128/mbio.00127-25] [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: 01/09/2025] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that has long been a significant threat to small ruminant productivity worldwide. Iron metabolism is vital to the host and the pathogen. However, the mechanism underlying host-PPRV interactions from the perspective of iron metabolism and iron-mediated membrane lipid peroxidation has not been reported thus far. In this study, we identified a novel host long-noncoding RNA (lncRNA), APR, that impairs PPRV infectivity by sponging miR-3955-5p, a negative microRNA (miRNA) that directly targets the gene encoding the ferritin-heavy chain 1 (FTH1) protein. Importantly, we demonstrated that PPRV infection causes aberrant cellular iron accumulation by increasing transferrin receptor (TFRC) expression and that iron accumulation induces reticulophagy and ferroptosis, which benefits PPRV replication. Moreover, PPRV infection enhanced the localization of cellular iron on the endoplasmic reticulum (ER) and caused ER membrane damage by promoting excess lipid peroxidation to induce reticulophagy. Interestingly, APR decreased PPRV infection-induced accumulation of intracellular Fe2+ via miR-3955-5p/FTH1 axis and ultimately inhibited reticulophagy and ferroptosis. Additionally, our results indicate that interferon regulatory factor 1 promotes APR transcription by positively regulating APR promoter activity after PPRV infection. Taken together, our findings revealed a new pattern of PPRV-host interactions, involving noncoding RNA regulation, iron metabolism, and iron-related membrane lipid peroxidation, which is critical for understanding the host defense against PPRV infection and the pathogenesis of PPRV.IMPORTANCEMany viruses have been demonstrated to engage in iron metabolism to facilitate their replication and pathogenesis. However, the mechanism by which PPRV interacts with host cells from the perspective of iron metabolism, or iron-mediated membrane lipid peroxidation, has not yet been reported. Our data provide the first direct evidence that PPRV infection induces aberrant iron accumulation to promote viral replication and reveal a novel host lncRNA, APR, as a regulator of iron accumulation by promoting FTH1 protein expression. In this study, PPRV infection increased cellular iron accumulation by increasing TFRC expression, and more importantly, iron overload increased viral infectivity as well as promoted ER membrane lipid peroxidation by enhancing the localization of cellular iron on the ER and ultimately induced ferroptosis and reticulophagy. Furthermore, a host factor, the lncRNA APR, was found to decrease cellular iron accumulation by sponging miR-3955-5p, which directly targets the gene encoding the FTH1 protein, thereby attenuating PPRV infection-induced ferroptosis and reticulophagy and inhibiting PPRV infection. Taken together, the results of the present study provide new insight into our understanding of host-PPRV interaction and pathogenesis from the perspective of iron metabolism and reveal potential targets for therapeutics against PPRV infection.
Collapse
Affiliation(s)
- Bo Wen
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Wenchi Chang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Lulu Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Daiyue Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
- China Institute of Veterinary Drug Control, Beijing, Beijing, China
| | - Lizhen Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Lei Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Yanzhao Xu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Jianhe Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Ke Ding
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, Beijing, China
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| | - Bo Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agriculture and Rural Affairs, Xi'an, Shaanxi, China
| |
Collapse
|
31
|
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
|
32
|
Xu Y, Ge M, Xu Y, Yin K. Ferroptosis: a novel perspective on tumor immunotherapy. Front Immunol 2025; 16:1524711. [PMID: 40260246 PMCID: PMC12009862 DOI: 10.3389/fimmu.2025.1524711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/11/2025] [Indexed: 04/23/2025] Open
Abstract
Ferroptosis is a novel form of programmed cell death characterized by iron-dependent accumulation of reactive oxygen species (ROS) and lipid peroxidation. The execution of ferroptosis is intricately linked to both iron and lipid metabolism. Intriguingly, iron and lipid metabolism are also pivotal for maintaining the physiological function of immune cells. Research has revealed that ferroptosis can potentiate the immunogenicity of tumor cells and engage in intricate interactions with immune cells. Certain ferroptosis inducers have the capacity to augment the efficacy of immunotherapy by modulating the tumor immune microenvironment. Ferroptosis holds immense potential in cancer immunotherapy and is anticipated to emerge as a novel therapeutic target in the future landscape of cancer treatment. In this review, we primarily delineate the ferroptosis signaling pathways and metabolic processes pertinent to immune cells, and further summarize the roles of ferroptosis in tumor-infiltrating immune cells. Ultimately, we anticipate further elucidation of the mechanisms of ferroptosis in immunotherapy and envision that strategies targeting ferroptosis and immunotherapy will be expeditiously applied in clinical oncology practice.
Collapse
Affiliation(s)
| | | | | | - Kai Yin
- Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
33
|
Lee WC, Dixon SJ. Mechanisms of ferroptosis sensitization and resistance. Dev Cell 2025; 60:982-993. [PMID: 40199240 DOI: 10.1016/j.devcel.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/08/2024] [Accepted: 02/05/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis is an iron-dependent and oxidative form of non-apoptotic cell death with roles in development, homeostasis, and disease. Ferroptosis sensitivity can vary between cells, often for reasons that are not well understood. In this perspective, we describe the core ferroptosis mechanism and outline how changes in iron, redox, and lipid metabolism can alter ferroptosis sensitivity. We propose the concept of a ferroptosis sensitivity-resistance continuum to describe how different intrinsic and extrinsic factors interact to push cells toward a more ferroptosis-sensitive or ferroptosis-resistant state, with effects on development and diseases such as cancer.
Collapse
Affiliation(s)
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
34
|
Chen Z, Niu K, Li M, Deng Y, Zhang J, Wei D, Wang J, Zhao Y. GCLC desuccinylation regulated by oxidative stress protects human cancer cells from ferroptosis. Cell Death Differ 2025:10.1038/s41418-025-01505-8. [PMID: 40188196 DOI: 10.1038/s41418-025-01505-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Tumor cells evolve strong antioxidant capacities to counteract the abnormal high level of reactive oxygen species (ROS) in the tumor microenvironment. Glutamate-cysteine ligase catalyzing subunit (GCLC) for synthesis of antioxidant glutathione (GSH) represents the key enzyme to maintain redox homeostasis of tumor cells, however, whether its activity is regulated by posttranslational modifications, such as succinylation, remains to be clarified. Here, we demonstrate the existence of succinylation modification on GCLC by in vitro and in vivo assays. NAD-dependent deacetylase Sirtuin-2 (SIRT2) serves as the desuccinylase and catalyzes GCLC desuccinylation at sites of K38, K126, and K326. Specifically, GCLC directly interacts with SIRT2, which can be substantially enhanced upon ROS treatment. This strengthened association results in GCLC desuccinylation and activation, consequently promoting GSH synthesis and rendering cancer cells resistant to ferroptosis induction. Depletion of SIRT2 decreases total GSH level and meanwhile increases the cellular susceptibility to ferroptosis, which can mostly be rescued by introducing wild-type GCLC, but not its 3K-E mutant. We further demonstrated that histone acetyltransferase P300 serves as the succinyltransferase of GCLC, and their association is remarkably decreased after ROS treatment. Thus, SIRT2-regulated GCLC succinylation represents an essential signaling axis for cancer cells to maintain their redox balance in coping with oxidative stress-induced ferroptosis.
Collapse
Affiliation(s)
- Zixiang Chen
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Kaifeng Niu
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Mengge Li
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuchun Deng
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ji Zhang
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Di Wei
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Jiaqi Wang
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongliang Zhao
- China National Center for Bioinformation, Beijing, China.
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
35
|
Huang Z, Zhou L, Liu B, Li X, Sang Y. Endoplasmic reticulum stress aggravates ferroptosis via PERK/ATF4/HSPA5 pathway in UUO-induced renal fibrosis. Front Pharmacol 2025; 16:1545972. [PMID: 40255561 PMCID: PMC12006179 DOI: 10.3389/fphar.2025.1545972] [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: 12/16/2024] [Accepted: 03/20/2025] [Indexed: 04/22/2025] Open
Abstract
Renal fibrosis, resulting from the transformation of damaged tubular epithelial cells (TECs), serves as a prevalent pathological condition observed in nearly all forms of advancing chronic kidney disease (CKD). Although crucial in fibrotic diseases, the association between endoplasmic reticulum stress (ERS) and ferroptosis remains incompletely elucidated. Herein, increased levels of heat shock protein family A member 5 (HSPA5), acting as a co-molecular in ERS and ferroptosis, along with EMT-associated alterations, including increased α-smooth muscle actin (α-SMA) and Col1a1 levels and decreased E-cad expression, were observed in fibrotic kidneys of Unilateral Ureteral Obstruction (UUO)-induced mouse models and TGF-β-induced EMT in HK-2 cells. The employment of ferrostatin-1 (Fer-1) improved these alterations and reversed TGF-β-induced EMT in vitro. More importantly, Inhibiting ERS by Tauroursodeoxycholate (TUDCA) reversed the alterations of ferroptosis, including GPX4 expression, reactive oxygen species (ROS) accumulation, iron overload, increased lipid peroxidation production, as well as EMT progression in vivo and in vitro. Whereas the overexpression of HSPA5 strikingly attenuated the inhibitory effects of TUDCA on ferroptosis and TGF-β-induced EMT in vitro. Mechanistically, Co-immunoprecipitation (Co-IP) tests showed that ATF4 engaged with and SUMOylated HSPA5 to trigger the HSPA5 signaling pathway in response to TGF-β. These findings illuminate that focusing on HSPA5 may present a promising therapeutic approach to enhance tubular epithelial cells' survival and alleviate the progression of CKD.
Collapse
Affiliation(s)
- Zhigang Huang
- Department of Urology, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Lihua Zhou
- Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Provincial Clinical Research Center for Natural Polymer Biological Liver, Wuhan Hubei, China
| | - Bin Liu
- Department of Urology, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Xiaoju Li
- Department of Urology, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Yu Sang
- Department of Urology, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| |
Collapse
|
36
|
Rutaganira FU, Coyle MC, Nguyen MHT, Hernandez I, Scopton AP, Dar AC, King N. A stress-responsive p38 signaling axis in choanoflagellates. RSC Chem Biol 2025:d4cb00122b. [PMID: 40226336 PMCID: PMC11984502 DOI: 10.1039/d4cb00122b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Animal kinases regulate cellular responses to environmental stimuli, including cell differentiation, migration, survival, and response to stress, but the ancestry of these functions is poorly understood. Choanoflagellates, the closest living relatives of animals, encode homologs of diverse animal kinases and have emerged as model organisms for reconstructing animal origins. However, efforts to identify key kinase regulators in choanoflagellates have been constrained by the limitations of currently available genetic tools. Here, we report on a framework that combines small molecule-driven kinase discovery with targeted genetics to reveal kinase function in choanoflagellates. To study the physiological roles of choanoflagellate kinases, we established two high-throughput platforms to screen the model choanoflagellate Salpingoeca rosetta with a curated library of human kinase inhibitors. We identified 95 diverse kinase inhibitors that disrupt S. rosetta cell proliferation. By focusing on one inhibitor, sorafenib, we identified a p38 kinase as a regulator of the heat shock response in S. rosetta. This finding reveals a conserved p38 function between choanoflagellates, animals, and fungi. Moreover, this study demonstrates that existing kinase inhibitors can serve as powerful tools to examine the ancestral roles of kinases that regulate modern animal development.
Collapse
Affiliation(s)
- Florentine U Rutaganira
- Department of Biochemistry, Stanford University School of Medicine Stanford CA 94305 USA
- Department of Developmental Biology, Stanford University School of Medicine Stanford CA 94305 USA
| | - Maxwell C Coyle
- Howard Hughes Medical Institute and the Department of Molecular and Cell Biology, University of California Berkeley CA 94720 USA
| | - Maria H T Nguyen
- Department of Biology, Stanford University Stanford CA 94305 USA
| | - Iliana Hernandez
- Department of Biochemistry, Stanford University School of Medicine Stanford CA 94305 USA
| | - Alex P Scopton
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai New York NY 10029 USA
| | - Arvin C Dar
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai New York NY 10029 USA
| | - Nicole King
- Howard Hughes Medical Institute and the Department of Molecular and Cell Biology, University of California Berkeley CA 94720 USA
| |
Collapse
|
37
|
Zhang Y, Su F, Zhu E, Sun Y, Kuang H, Wang Q. A systematical review on traditional Chinese medicine treating chronic diseases via regulating ferroptosis from the perspective of experimental evidence and clinical application. CHINESE HERBAL MEDICINES 2025; 17:246-260. [PMID: 40256717 PMCID: PMC12009076 DOI: 10.1016/j.chmed.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/12/2024] [Accepted: 01/20/2025] [Indexed: 04/22/2025] Open
Abstract
Ferroptosis is a unique regulated form of cell death that is distinct from apoptosis, necrosis, and other well-characterized regulated cell death types, and plays an important role in the occurrence and development of chronic metabolic diseases, including diabetes, hypertension, hyperlipidemia, and non-alcoholic steatohepatitis. Recently, increasing evidence has supported traditional Chinese medicine (TCM) as a new hot spot for the treatment of chronic metabolic diseases by mediating ferroptosis. Unfortunately, few systematic reviews have described the importance of TCM in treating chronic metabolic diseases through the ferroptosis pathway. In the current review, the mechanism of ferroptosis and the roles of ferroptosis in chronic metabolic diseases are summarized. Additionally, this review illustrates that the regulation of ferroptosis by TCM could be an effective approach for treating chronic metabolic diseases based on experimental evidence and clinical application. In summary, this work will improve the understanding of ferroptosis and the ability of TCM to regulate ferroptosis in chronic metabolic diseases, thereby promoting the development and application of natural TCM.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Fazhi Su
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Enlin Zhu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Qiuhong Wang
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150040, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 511400, China
| |
Collapse
|
38
|
Prabhune NM, Ameen B, Prabhu S. Therapeutic potential of synthetic and natural iron chelators against ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3527-3555. [PMID: 39601820 DOI: 10.1007/s00210-024-03640-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Ferroptosis, a regulated form of cell death, is characterized by iron accumulation that results in the production of reactive oxygen species. This further causes lipid peroxidation and damage to the cellular components, eventually culminating into oxidative stress. Recent studies have highlighted the pivotal role of ferroptosis in the pathophysiological development and progression of various diseases such as β-thalassemia, hemochromatosis, and neurodegenerative disorders like AD and PD. Extensive efforts are in progress to understand the molecular mechanisms governing the role of ferroptosis in these conditions, and chelation therapy stands out as a potential approach to mitigate ferroptosis and its related implications in their development. There are currently both synthetic and natural iron chelators that are being researched for their potential as ferroptosis inhibitors. While synthetic chelators are relatively well-established and studied, their short plasma half-life and toxic side effects necessitate the exploration and identification of natural products that can act as efficient and safe iron chelators. In this review, we comprehensively discuss both synthetic and natural iron chelators as potential therapeutic strategies against ferroptosis-induced pathologies.
Collapse
Affiliation(s)
- Nupura Manish Prabhune
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Bilal Ameen
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sudharshan Prabhu
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
39
|
Yang Z, Yuan Y, Niu Y, Zuo D, Liu W, Li K, Shi Y, Qiu Z, Li K, Lin Z, Zhong C, Huang Z, He W, Guan X, Yuan Y, Zeng W, Qiu J, Li B. Regulatory factor X1 promotes sorafenib-induced ferroptosis in hepatocellular carcinoma by transcriptional regulation of BECN1. Cell Oncol (Dordr) 2025; 48:505-522. [PMID: 39652303 PMCID: PMC11996997 DOI: 10.1007/s13402-024-01017-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Sorafenib is a commonly used first-line kinase-targeted drug for advanced hepatocellular carcinoma (HCC) patients suffering from limited efficacy. Emerging evidence indicates that sorafenib exerts anti-cancer activity through the induction of ferroptosis in HCC cells, but the underlying mechanism is still unclear. METHODS The whole transcriptome sequencing and bioinformatics analysis were used to screen for target genes. The expression and subcellular localization of regulatory factor X1 (RFX1) were determined through immunohistochemistry, immunofluorescence, PCR and western blot analyses. The impact of RFX1 on HCC cell growth was assessed using CCK8, colony formation assays, cell death assays, and animal experiments. Glutathione measurement, iron assay and lipid peroxidation detection assays were performed to investigate ferroptosis of HCC cells. The regulatory mechanism of RFX1 in HCC was investigated by sgRFX1, co-IP, ChIP and luciferase experiments. Immunohistochemical and survival analyses were performed to examine the prognostic significance of RFX1 in HCC. RESULTS In this study, we found that RFX1 promote ferroptosis in HCC cells. Further, we showed that sorafenib induces cell death through RFX1-mediated ferroptosis in HCC cells. The enhancing effect of RFX1 on HCC cell ferroptosis is largely dependent on inhibition of cystine/glutamate antiporter (system Xc-) activity through the BECN-SLC7A11 axis, where RFX1 directly binds to the promoter region of BECN1 and upregulates BECN1 expression. In addition, a STAT3-RFX1-BECN1 signalling loop was found to promote RFX1 expression in HCC cells. CONCLUSIONS Our study reveals a novel mechanism underlying sorafenib-induced HCC cell death.
Collapse
Affiliation(s)
- Zhiwen Yang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Yichuan Yuan
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Yi Niu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Dinglan Zuo
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wenwu Liu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Kai Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Yunxing Shi
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Zhiyu Qiu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Keren Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Zhu Lin
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Chengrui Zhong
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Zhenkun Huang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Wei He
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Xinyuan Guan
- Department of Clinical Oncology, State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Yunfei Yuan
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China
| | - Weian Zeng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China.
| | - Jiliang Qiu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China.
| | - Binkui Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, PR China.
| |
Collapse
|
40
|
Hu B, Yin Y, Zhang B, Li S, Li K, Zhou Y, Huang Q. Villin-1 regulates ferroptosis in colorectal cancer progression. FEBS J 2025; 292:1710-1725. [PMID: 39658274 DOI: 10.1111/febs.17350] [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: 04/16/2024] [Revised: 08/01/2024] [Accepted: 09/13/2024] [Indexed: 12/12/2024]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. Despite extensive research, the mechanistic underpinnings driving CRC progression remain largely unknown. As a fundamental component of the brush border cytoskeleton, villin-1 (VIL1) acts as a marker for intestinal cell differentiation and maturation. Through a comprehensive transcriptomics analysis of eight studies (total sample: n = 1952), we consistently observed significant upregulation of VIL1 expression in CRC tumors compared with adjacent normal tissue. In our independent cohort, this notable upregulation has been further validated at both mRNA and protein levels in colon tumor tissues, relative not only to adjacent normal tissue but also to normal controls. Our data show that VIL1 promotes proliferation and migration while inhibiting apoptosis. Conversely, knockout of VIL1 suppresses proliferation and migration while inducing apoptosis. Mechanistically, we reveal that knocking out VIL1 activates ferroptosis and inhibits the migration of CRC cells, while overexpressing VIL1 yields the opposite effects, and vice versa. Additionally, VIL1 binds to Nuclear factor NF-kappa-B p105 subunit (NF-κB) and controls NF-κB expression. In vivo, overexpressing VIL1 inhibits ferroptosis, and induces the expression of NF-κB and lipocalin 2 (LCN2), thereby promoting CRC tumor growth. Thus, we have identified the VIL1/NF-κB axis as a pivotal regulator of CRC progression through ferroptosis modulation, unveiling VIL1 as a promising therapeutic target for CRC treatment via ferroptosis. Our study offers novel avenues for exploring the therapeutic potential of ferroptosis in CRC management, emphasizing the high potential of VIL1 in regulating colorectal tumorigenesis.
Collapse
Affiliation(s)
- Bangli Hu
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Yixin Yin
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Birong Zhang
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Siqi Li
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Kezhi Li
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - You Zhou
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Qinghua Huang
- Department of Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Department of Breast Surgery, Wuzhou Red Cross Hospital, Wuzhou, China
| |
Collapse
|
41
|
Zheng J, Conrad M. Ferroptosis: when metabolism meets cell death. Physiol Rev 2025; 105:651-706. [PMID: 39661331 DOI: 10.1152/physrev.00031.2024] [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: 08/08/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
We present here a comprehensive update on recent advancements in the field of ferroptosis, with a particular emphasis on its metabolic underpinnings and physiological impacts. After briefly introducing landmark studies that have helped to shape the concept of ferroptosis as a distinct form of cell death, we critically evaluate the key metabolic determinants involved in its regulation. These include the metabolism of essential trace elements such as selenium and iron; amino acids such as cyst(e)ine, methionine, glutamine/glutamate, and tryptophan; and carbohydrates, covering glycolysis, the citric acid cycle, the electron transport chain, and the pentose phosphate pathway. We also delve into the mevalonate pathway and subsequent cholesterol biosynthesis, including intermediate metabolites like dimethylallyl pyrophosphate, squalene, coenzyme Q (CoQ), vitamin K, and 7-dehydrocholesterol, as well as fatty acid and phospholipid metabolism, including the biosynthesis and remodeling of ester and ether phospholipids and lipid peroxidation. Next, we highlight major ferroptosis surveillance systems, specifically the cyst(e)ine/glutathione/glutathione peroxidase 4 axis, the NAD(P)H/ferroptosis suppressor protein 1/CoQ/vitamin K system, and the guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin/dihydrofolate reductase axis. We also discuss other potential anti- and proferroptotic systems, including glutathione S-transferase P1, peroxiredoxin 6, dihydroorotate dehydrogenase, glycerol-3-phosphate dehydrogenase 2, vitamin K epoxide reductase complex subunit 1 like 1, nitric oxide, and acyl-CoA synthetase long-chain family member 4. Finally, we explore ferroptosis's physiological roles in aging, tumor suppression, and infection control, its pathological implications in tissue ischemia-reperfusion injury and neurodegeneration, and its potential therapeutic applications in cancer treatment. Existing drugs and compounds that may regulate ferroptosis in vivo are enumerated.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, Garching, Germany
| |
Collapse
|
42
|
Varlamova EG. Roles of selenium-containing glutathione peroxidases and thioredoxin reductases in the regulation of processes associated with glioblastoma progression. Arch Biochem Biophys 2025; 766:110344. [PMID: 39956249 DOI: 10.1016/j.abb.2025.110344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Glioblastoma remains the most common and aggressive primary tumor of the central nervous system in adults. Current treatment options include standard surgical resection combined with radiation/chemotherapy, but such protocol most likely only delays the inevitable. Therefore, the problem of finding therapeutic targets to prevent the occurrence and development of this severe oncological disease is currently acute. It is known that the functions of selenoproteins in the regulation of carcinogenesis processes are not unambiguous. Either they exhibit cytotoxic activity on cancer cells, or cytoprotective. A special place in the progression of oncological diseases of various etiologies is occupied by proteins of the thioredoxin and glutathione systems. These are two cellular antioxidant systems that regulate redox homeostasis, counteracting the increased production of reactive oxygen species in cells. The review reflects the latest data on the role of key enzymes of these redox systems in the regulation of processes associated with the progression of glioblastoma. A thorough consideration of these issues will expand fundamental knowledge about the functions of selenium-containing thioredoxin reductases and glutathione peroxidases in the therapy of glioblastomas and provide an understanding of the prospects for the treatment of this aggressive oncological disease.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", St. Institutskaya 3, Pushchino, 142290, Russia.
| |
Collapse
|
43
|
Fang Y, Chen H, Liu Y, Jiang K, Qian Y, Wei J, Fu D, Yang H, Dai S, Jin T, Bu T, Ding K. NUPR1 Promotes Radioresistance in Colorectal Cancer Cells by Inhibiting Ferroptosis. J Cell Mol Med 2025; 29:e70519. [PMID: 40176685 PMCID: PMC11965884 DOI: 10.1111/jcmm.70519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 01/20/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
Radioresistance is a major clinical challenge and the underlying mechanism has not been thoroughly elucidated. In this study, a radioresistant (RR) cell line is established to explore the transcriptomic signatures of radioresistance in colorectal cancer (CRC). KEGG enriched pathway analysis demonstrated that ferroptosis is inactivated in RR cells. Further detection confirmed that radiotherapy can promote ferroptosis, and ferroptosis inactivation is one of the hallmarks of radioresistance in CRC. What's more, induction of ferroptosis can restore the radiosensitivity of CRC cells. Then, we performed RNA sequencing to compare gene expression between parental and RR cells, and cells pretreated with or without RSL3. Via high-throughput screening, NUPR1 was identified as a potential candidate for ferroptosis-mediated radioresistance in CRC. CRC cells can acquire radiation resistance by NUPR1-mediated ferroptosis suppression in the NUPR1-overexpressing cell line. More importantly, ZZW-115, an NUPR1 inhibitor, can sensitise RR cells to radiotherapy. Overall, our findings identify ferroptosis inactivation linked with resistance to radiotherapy. Besides, NUPR1 can promote radiation resistance by inhibiting ferroptosis, and targeting NUPR1 may be a potential strategy to relieve radioresistance associated with ferroptosis in CRC.
Collapse
Affiliation(s)
- Yimin Fang
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Haiyan Chen
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
- Department of Radiation Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouZhejiangChina
| | - Yunhua Liu
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Kai Jiang
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Yucheng Qian
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Jingsun Wei
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Dongliang Fu
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Hang Yang
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Siqi Dai
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Tian Jin
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Tongtong Bu
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology(Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), the Second Affiliated Hospital, Zhejiang University School of MedicineZhejiangHangzhouChina
- Zhejiang Provincial Clinical Research Center for CANCERHangzhouChina
- Cancer Center of Zhejiang UniversityHangzhouChina
| |
Collapse
|
44
|
Lan J, Cai D, Gou S, Bai Y, Lei H, Li Y, Chen Y, Zhao Y, Shen J, Wu X, Li M, Chen M, Li X, Sun Y, Gu L, Li W, Wang F, Cho CH, Zhang Y, Zheng X, Xiao Z, Du F. The dynamic role of ferroptosis in cancer immunoediting: Implications for immunotherapy. Pharmacol Res 2025; 214:107674. [PMID: 40020885 DOI: 10.1016/j.phrs.2025.107674] [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: 12/19/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/03/2025]
Abstract
Currently, cancer immunotherapy strategies are primarily formulated based on the patient's present condition, representing a "static" treatment approach. However, cancer progression is inherently "dynamic," as the immune environment is not fixed but undergoes continuous changes. This dynamism is characterized by the ongoing interactions between tumor cells and immune cells, which ultimately lead to alterations in the tumor immune microenvironment. This process can be effectively elucidated by the concept of cancer immunoediting, which divides tumor development into three phases: "elimination," "equilibrium," and "escape." Consequently, adjusting immunotherapy regimens based on these distinct phases may enhance patient survival and improve prognosis. Targeting ferroptosis is an emerging area in cancer immunotherapy, and our findings reveal that the antioxidant systems associated with ferroptosis possess dual roles, functioning differently across the three phases of cancer immunoediting. Therefore, this review delve into the dual role of the ferroptosis antioxidant system in tumor development and progression. It also propose immunotherapy strategies targeting ferroptosis at different stages, ultimately aiming to illuminate the significant implications of targeting ferroptosis at various phases for cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarui Lan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Dan Cai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Shuang Gou
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Yulin Bai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China
| | - Huaqing Lei
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yan Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Zhang
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China
| | - Xin Zheng
- Department of Oncology, Luzhou People's Hospital, Luzhou, Sichuan 646000, China.
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646600, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan 646000, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan 646600, China.
| |
Collapse
|
45
|
Yang X, Wu H, Liu D, Zhou G, Zhang D, Yang Q, Liu Y, Li Y. The link between ferroptosis and autophagy in myocardial ischemia/reperfusion injury: new directions for therapy. J Cardiovasc Transl Res 2025; 18:408-423. [PMID: 39885084 DOI: 10.1007/s12265-025-10590-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
Myocardial ischemia/reperfusion (I/R)-induced cell death, such as autophagy and ferroptosis, is a major contributor to cardiac injury. Regulating cell death may be key to mitigating myocardial ischemia/reperfusion injury (MI/RI). Autophagy is a crucial physiological process involving cellular self-digestion and compensation, responsible for degrading excess or malfunctioning long-lived proteins and organelles. During MI/RI, autophagy plays both "survival" and "death" roles. A growing body of research indicates that ferroptosis is a type of autophagy-dependent cell death. This article provides a comprehensive review of the functions of autophagy and ferroptosis in MI/RI, as well as the molecules mediating their interaction. Understanding the link between autophagy and ferroptosis may offer new therapeutic directions for MI/RI, bearing significant clinical implications.
Collapse
Affiliation(s)
- Xiaoting Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| | - Hui Wu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China.
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China.
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China.
| | - Di Liu
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huang Shi, HuBei Province, China
| | - Gang Zhou
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Dong Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Qingzhuo Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yanfang Liu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| | - Yi Li
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| |
Collapse
|
46
|
Xia X, Kong C, Zhao X, Zhao K, Shi N, Jiang J, Li P. The complexities of cell death mechanisms: a new perspective in systemic sclerosis therapy. Apoptosis 2025; 30:636-651. [PMID: 39924583 DOI: 10.1007/s10495-025-02082-4] [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] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
Systemic sclerosis, also termed scleroderma, is a severe and debilitating autoimmune disease characterized by fibrosis, an aberrant immune response, and vascular dysfunction. Cell death is essential to the body's continued normal development as it removes old or damaged cells. This process is governed by several mechanisms, including programmed cell death through apoptosis, necrosis, and pyroptosis, as well as metabolic processes, such as ferroptosis and cuproptosis. This review describes the signaling pathways associated with each form of cell death, examining the linkages between these pathways, and discussing how the dysregulation of cell death processes is involved in the development of autoimmune disorders such as systemic sclerosis. Existing and promising therapeutic strategies aimed at restoring the balance of cell death in systemic sclerosis and other autoimmune disorders are also emphasized.
Collapse
Affiliation(s)
- Xue Xia
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Chenfei Kong
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Xiaoming Zhao
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Kelin Zhao
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Naixu Shi
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, 130033, China.
| | - Ping Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China.
| |
Collapse
|
47
|
Zhang L, Li Y, Qian Y, Xie R, Peng W, Zhou W. Advances in the Development of Ferroptosis-Inducing Agents for Cancer Treatment. Arch Pharm (Weinheim) 2025; 358:e202500010. [PMID: 40178208 DOI: 10.1002/ardp.202500010] [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: 01/04/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 04/05/2025]
Abstract
Cancer is the main leading cause of death worldwide and poses a great threat to human life and health. Although pharmacological treatment with chemotherapy and immunotherapy is the main therapeutic strategy for cancer patients, there are still many shortcomings during the treatment such as incomplete killing of cancer cells and development of drug resistance. Emerging evidence indicates the promise of inducing ferroptosis for cancer treatment, particularly for eliminating aggressive malignancies that are resistant to conventional therapies. This review covers recent advances in important regulatory targets in the ferroptosis metabolic pathway and ferroptosis inducers (focusing mainly on the last 3 years) to delineate their design, mechanisms of action, and anticancer applications. To date, many compounds, including inhibitors, degraders, and active molecules from traditional Chinese medicine, have been demonstrated to have ferroptosis-inducing activity by targeting the different biomolecules in the ferroptosis pathway. However, strictly defined ferroptosis inducers have not yet been approved for clinical use; therefore, the discovery of new highly active, less toxic, and selective compounds remains the goal of further research in the coming years.
Collapse
Affiliation(s)
- Li Zhang
- Maternal and Child Health Department, Shaoxing Maternity and Child Health Care Hospital, Shaoxing, Zhejiang Province, China
| | - Yulong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yufeng Qian
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, China
| | - Ruliang Xie
- Jiangsu Institute of Marine Resources Development, Jiangsu Ocean University, Lianyungang, Jiangsu Province, China
| | - Wei Peng
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, China
| | - Wen Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
48
|
Kunishige R, Noguchi Y, Okamoto N, Li L, Ono A, Murata M, Kano F. Protein covariation networks for elucidating ferroptosis inducer mechanisms and potential synergistic drug targets. Commun Biol 2025; 8:480. [PMID: 40164758 PMCID: PMC11958834 DOI: 10.1038/s42003-025-07886-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
In drug development, systematically characterizing a compound's mechanism of action (MoA), including its direct targets and effector proteins, is crucial yet challenging. Network-based approaches, unlike those focused solely on direct targets, effectively detect a wide range of cellular responses elicited by compounds. This study applied protein covariation network analysis, leveraging quantitative, morphological, and localization features from immunostained microscopic images, to elucidate the MoA of AX-53802, a novel ferroptosis inducer. From the candidate targets extracted through network analysis, GPX4 was verified as the direct target by validation experiments. Additionally, aggregates involving GPX4, TfR1, and F-actin were observed alongside iron reduction, suggesting a ferroptosis defense mechanism. Furthermore, combination therapies targeting GPX4 and FAK/Src were found to enhance cancer cell death, and MDM2, ezrin, and cortactin were identified as potential ferroptosis inhibitor targets. These findings highlight the effectiveness of network-based approaches in uncovering a compound's MoA and developing combination therapies for cancer.
Collapse
Affiliation(s)
- Rina Kunishige
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Yoshiyuki Noguchi
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
- International Research Center for Neurointelligence, Institutes for Advanced Study, The University of Tokyo, Tokyo, Japan
| | | | - Lei Li
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Akito Ono
- Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Masayuki Murata
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Fumi Kano
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan.
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan.
- Cell Biology Center, Institute of Integrated Research, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan.
| |
Collapse
|
49
|
Jiang C, Yan Y, Long T, Xu J, Chang C, Kang M, Wang X, Chen Y, Qiu J. Ferroptosis: a potential therapeutic target in cardio-cerebrovascular diseases. Mol Cell Biochem 2025:10.1007/s11010-025-05262-7. [PMID: 40148662 DOI: 10.1007/s11010-025-05262-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Cardio-cerebrovascular diseases (CCVDs) are the leading cause of global mortality, yet effective treatment options remain limited. Ferroptosis, a novel form of regulated cell death, has emerged as a critical player in various CCVDs, including atherosclerosis, myocardial infarction, ischemia-reperfusion injury, cardiomyopathy, and ischemic/hemorrhagic strokes. This review highlights the core mechanisms of ferroptosis, its pathological implications in CCVDs, and the therapeutic potential of targeting this process. Additionally, it explores the role of Chinese herbal medicines (CHMs) in mitigating ferroptosis, offering novel therapeutic strategies for CCVDs management. Ferroptosis is regulated by several key pathways. The GPX4-GSH-System Xc- axis is central to ferroptosis execution, involving GPX4 using GSH to neutralize lipid peroxides, with system Xc- being crucial for GSH synthesis. The NAD(P)H/FSP1/CoQ10 axis involves FSP1 regenerating CoQ10 via NAD(P)H, inhibiting lipid peroxidation independently of GPX4. Lipid peroxidation, driven by PUFAs and enzymes like ACSL4 and LPCAT3, and iron metabolism, regulated by proteins like TfR1 and ferritin, are also crucial for ferroptosis. Inhibiting ferroptosis shows promise in managing CCVDs. In atherosclerosis, ferroptosis inhibitors reduce iron accumulation and lipid peroxidation. In myocardial infarction, inhibitors protect cardiomyocytes by preserving GPX4 and SLC7A11 levels. In ischemia-reperfusion injury, targeting ferroptosis reduces myocardial and cerebral damage. In diabetic cardiomyopathy, Nrf2 activators alleviate oxidative stress and iron metabolism irregularities. CHMs offer natural compounds that mitigate ferroptosis. They possess antioxidant properties, chelate iron, and modulate signaling pathways like Nrf2 and AMPK. For example, Salvia miltiorrhiza and Astragalus membranaceus reduce oxidative stress, while some CHMs chelate iron, reducing its availability for ferroptosis. In conclusion, ferroptosis plays a pivotal role in CCVDs, and targeting it offers novel therapeutic avenues. CHMs show promise in reducing ferroptosis and improving patient outcomes. Future research should explore combination therapies and further elucidate the molecular interactions in ferroptosis.
Collapse
Affiliation(s)
- Chenlong Jiang
- School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Yang Yan
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Tianlin Long
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Jiawei Xu
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Cuicui Chang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
- Department of Cardiology, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China
| | - Meili Kang
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China
| | - Xuanqi Wang
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| | - Yuhua Chen
- Department of Medical Science Research Center, Xi'an Peihua University, No. 888 Changning Road, Xi'an, 710125, Shaanxi, China.
- Department of Neurosurgery, Bijie Traditional Chinese Medicine Hospital, Bijie, 551700, China.
- School of Life and Health Science, Hainan University, No. 58 People's Avenue, Haikou, 570100, Hainan, China.
| | - Junlin Qiu
- Department of Cardiology, First Hospital of Northwestern University, Northwest University, No. 512 Xianning East Road, Xi'an, 710043, Shaanxi, China.
| |
Collapse
|
50
|
Sanchez KL, Kim J, White JB, Tolan A, Rajagopal NP, Anderson DW, Shin AN, Shin SD, Currais A, Soriano-Castell D, Maher P, Soriano S. Evidence of Oxytosis/Ferroptosis in Niemann-Pick Disease Type C. Int J Mol Sci 2025; 26:2915. [PMID: 40243519 PMCID: PMC11988824 DOI: 10.3390/ijms26072915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Niemann-Pick Disease Type C (NPC) is a hereditary neurodegenerative disease characterized by selective cell vulnerability, particularly affecting cerebellar anterior Purkinje neurons. These neurons exhibit a distinctive pattern of degeneration due to the loss of NPC1 and/or NPC2 protein function, progressively extending towards posterior cerebellar regions. Our study aimed to explore the early factors influencing this selective vulnerability of anterior Purkinje neurons in NPC. Oxytosis/ferroptosis, a novel form of regulated cell death, has been implicated in neurodegenerative diseases, with its inhibition showing promising therapeutic potential. Our laboratory has previously identified parallels between NPC cellular pathology and ferroptotic markers, including elevated levels of lipid peroxidation and iron, mitochondrial dysfunction, and Ca2+ dyshomeostasis. However, whether oxytosis/ferroptosis underlies NPC cellular pathology remains unexplored. We hypothesize that loss of NPC1 function increases vulnerability to ferroptosis and that anti-ferroptotic compounds will reverse NPC cellular pathology. Through bioinformatic analyses of pre-symptomatic Npc1-/- Purkinje neurons and in vitro studies using primary dermal fibroblasts derived from NPC patients, we provide evidence suggesting that oxytosis/ferroptosis may play a pathogenic role in NPC. These findings highlight the potential of anti-ferroptotic compounds as a promising therapeutic strategy to mitigate neurodegeneration in NPC and potentially other related disorders.
Collapse
Affiliation(s)
- Kayla L. Sanchez
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Jeanyoung Kim
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Jacob B. White
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Andrew Tolan
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Naren P. Rajagopal
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Douglas W. Anderson
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Alexandra N. Shin
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Samuel D. Shin
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| | - Antonio Currais
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - David Soriano-Castell
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Pamela Maher
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA; (A.C.); (D.S.-C.)
| | - Salvador Soriano
- Department of Pathology and Human Anatomy, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.L.S.); (J.K.); (J.B.W.); (N.P.R.); (D.W.A.); (A.N.S.); (S.D.S.)
| |
Collapse
|