1
|
Xiao Y, He M, Zhang X, Yang M, Yuan Z, Yao S, Qin Y. Research progress on the mechanism of tumor cell ferroptosis regulation by epigenetics. Epigenetics 2025; 20:2500949. [PMID: 40327848 PMCID: PMC12064064 DOI: 10.1080/15592294.2025.2500949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer remains a significant barrier to human longevity and a leading cause of mortality worldwide. Despite advancements in cancer therapies, challenges such as cellular toxicity and drug resistance to chemotherapy persist. Regulated cell death (RCD), once regarded as a passive process, is now recognized as a programmed mechanism with distinct biochemical and morphological characteristics, thereby presenting new therapeutic opportunities. Ferroptosis, a novel form of RCD characterized by iron-dependent lipid peroxidation and unique mitochondrial damage, differs from apoptosis, autophagy, and necroptosis. It is driven by reactive oxygen species (ROS)-induced lipid peroxidation and is implicated in tumorigenesis, anti-tumor immunity, and resistance, particularly in tumors undergoing epithelial-mesenchymal transition. Moreover, ferroptosis is associated with ischemic organ damage, degenerative diseases, and aging, regulated by various cellular metabolic processes, including redox balance, iron metabolism, and amino acid, lipid, and glucose metabolism. This review focuses on the role of epigenetic factors in tumor ferroptosis, exploring their mechanisms and potential applications in cancer therapy. It synthesizes current knowledge to provide a comprehensive understanding of epigenetic regulation in tumor cell ferroptosis, offering insights for future research and clinical applications.
Collapse
Affiliation(s)
- Yuyang Xiao
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mengyang He
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xupeng Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Meng Yang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhangchi Yuan
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shanhu Yao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Medical Information Research, Central South University, Changsha, Hunan, China
| | - Yuexiang Qin
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Kato K, Yasui H, Sato-Akaba H, Emoto MC, Fujii HG, Kmiec MM, Kuppusamy P, Nagane M, Yamashita T, Inanami O. Non-invasive electron paramagnetic resonance imaging detects tumor redox imbalance induced by ferroptosis. Redox Rep 2025; 30:2454887. [PMID: 39836064 PMCID: PMC11753017 DOI: 10.1080/13510002.2025.2454887] [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] [Indexed: 01/22/2025] Open
Abstract
Targeting ferroptosis, cell death caused by the iron-dependent accumulation of lipid peroxides, and disruption of the redox balance are promising strategies in cancer therapy owing to the physiological characteristics of cancer cells. However, the detection of ferroptosis using in vivo imaging remains challenging. We previously reported that redox maps showing the reduction power per unit time of implanted tumor tissues via non-invasive redox imaging using a novel, compact, and portable electron paramagnetic resonance imaging (EPRI) device could be compared with tumor tissue sections. This study aimed to apply the EPRI technique to the in vivo detection of ferroptosis. Notably, redox maps reflecting changes in the redox status of tumors induced by the ferroptosis-inducing agent imidazole ketone erastin (IKE) were compared with the immunohistochemical images of 4-hydroxynonenal (4-HNE) in tumor tissue sections. Our comparison revealed a negative correlation between the reducing power of tumor tissue and the number of 4-HNE-positive cells. Furthermore, the control and IKE-treated groups exhibited significantly different distributions on the correlation map. Therefore, redox imaging using EPRI may contribute to the non-invasive detection of ferroptosis in vivo.
Collapse
Affiliation(s)
- Kazuhiro Kato
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Hideo Sato-Akaba
- Department of Electrical and Electronic Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, Japan
| | - Miho C. Emoto
- Department of Clinical Laboratory Science, School of Medical Technology, Health Sciences University of Hokkaido, Sapporo, Japan
| | - Hirotada G. Fujii
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari, Japan
| | - Maciej M. Kmiec
- Departments of Radiology and Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Periannan Kuppusamy
- Departments of Radiology and Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Masaki Nagane
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Tadashi Yamashita
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
3
|
Zheng Q, Wang D, Lin R, Xu W. Pyroptosis, ferroptosis, and autophagy in spinal cord injury: regulatory mechanisms and therapeutic targets. Neural Regen Res 2025; 20:2787-2806. [PMID: 39101602 PMCID: PMC11826477 DOI: 10.4103/nrr.nrr-d-24-00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/24/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Regulated cell death is a form of cell death that is actively controlled by biomolecules. Several studies have shown that regulated cell death plays a key role after spinal cord injury. Pyroptosis and ferroptosis are newly discovered types of regulated cell deaths that have been shown to exacerbate inflammation and lead to cell death in damaged spinal cords. Autophagy, a complex form of cell death that is interconnected with various regulated cell death mechanisms, has garnered significant attention in the study of spinal cord injury. This injury triggers not only cell death but also cellular survival responses. Multiple signaling pathways play pivotal roles in influencing the processes of both deterioration and repair in spinal cord injury by regulating pyroptosis, ferroptosis, and autophagy. Therefore, this review aims to comprehensively examine the mechanisms underlying regulated cell deaths, the signaling pathways that modulate these mechanisms, and the potential therapeutic targets for spinal cord injury. Our analysis suggests that targeting the common regulatory signaling pathways of different regulated cell deaths could be a promising strategy to promote cell survival and enhance the repair of spinal cord injury. Moreover, a holistic approach that incorporates multiple regulated cell deaths and their regulatory pathways presents a promising multi-target therapeutic strategy for the management of spinal cord injury.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Weihong Xu
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
4
|
Guo W, Duan Z, Wu J, Zhou BP. Epithelial-mesenchymal transition promotes metabolic reprogramming to suppress ferroptosis. Semin Cancer Biol 2025; 112:20-35. [PMID: 40058616 DOI: 10.1016/j.semcancer.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular de-differentiation process that provides cells with the increased plasticity and stem cell-like traits required during embryonic development, tissue remodeling, wound healing and metastasis. Morphologically, EMT confers tumor cells with fibroblast-like properties that lead to the rearrangement of cytoskeleton (loss of stiffness) and decrease of membrane rigidity by incorporating high level of poly-unsaturated fatty acids (PUFA) in their phospholipid membrane. Although large amounts of PUFA in membrane reduces rigidity and offers capabilities for tumor cells with the unbridled ability to stretch, bend and twist in metastasis, these PUFA are highly susceptible to lipid peroxidation, which leads to the breakdown of membrane integrity and, ultimately results in ferroptosis. To escape the ferroptotic risk, EMT also triggers the rewiring of metabolic program, particularly in lipid metabolism, to enforce the epigenetic regulation of EMT and mitigate the potential damages from ferroptosis. Thus, the interplay among EMT, lipid metabolism, and ferroptosis highlights a new layer of intricated regulation in cancer biology and metastasis. Here we summarize the latest findings and discuss these mutual interactions. Finally, we provide perspectives of how these interplays contribute to cellular plasticity and ferroptosis resistance in metastatic tumor cells that can be explored for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Wenzheng Guo
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Zhibing Duan
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Jingjing Wu
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States
| | - Binhua P Zhou
- Departments of Molecular and Cellular Biochemistry, and the Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40506, United States.
| |
Collapse
|
5
|
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] [MESH Headings] [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
|
6
|
Liu X, Long G, Wang Q, Wang J, Chen X, Zhou Y, You K, Li H, Jiang Y, Zhang Z, Du X, Han Y. ANG secretion predisposes endothelial cells toward angiogenesis in FIN56-induced ferroptotic hepatocellular carcinoma via the BMP6/ID1 signaling pathway. Free Radic Biol Med 2025; 238:S0891-5849(25)00778-6. [PMID: 40541804 DOI: 10.1016/j.freeradbiomed.2025.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 06/08/2025] [Accepted: 06/17/2025] [Indexed: 06/22/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) poses significant challenges due to its high malignancy and limited treatment options. FIN56 has emerged as a potent inducer of ferroptosis, yet its precise mechanism of action in HCC remains elusive. METHODS Ferroptosis induction by FIN56 in HCC cells was assessed by quantifying malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), reactive oxygen species (ROS), glutathione (GSH) and Liperfluo levels, as well as evaluating changes in mitochondrial morphology.Additionally, cell proliferation and migration assays were performed to assess the functional impact of FIN56 on HCC cells. Tube formation and sprouting assays were performed using human umbilical vein endothelial cells (HUVECs). Proteomics analysis and immunosorbent assay (ELISA) identified angiogenin (ANG) as a secreted protein in the supernatant of HCC cells. Furthermore, both xenograft and syngeneic tumor models were established to investigate the potential impact of FIN56-induced ferroptosis on the tumor microenvironment. RESULTS RNA sequencing analysis of FIN56-treated HCC cells identified differentially expressed genes mainly associated with ferroptosis, cell proliferation, and migration. FIN56 effectively induced ferroptosis in HCC cells, while simultaneously inhibiting their proliferation and migration. RNA sequencing of HUVECs exposed to conditioned medium(CM) from FIN56-treated HCC cells (FIN56-CM) showed significant alterations in endothelial cell growth and gene expression. Further experiments revealed that FIN56-treated HCC cells secreted substantial levels of ANG after 12-18h, which activated the BMP6/ID1 signaling axis in endothelial cells, thereby enhancing their tube formation. Notably, the BMP signaling inhibitor LDN214117 partially suppressed ANG-mediated effects on endothelial cells. Finally, both ferroptosis and pro-angiogenic effects of FIN56 were confirmed in xenograft tumor model. CONCLUSION FIN56 induces ferroptosis in HCC cells, leading to the secretion of ANG. The secreted ANG acts as a critical signaling molecule, activating the BMP6/ID1 pathway in HUVECs and contributing to tumor microenvironment regulation.
Collapse
Affiliation(s)
- Xin Liu
- Department of oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Gang Long
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin 150007, China
| | - Qingwen Wang
- Department of Urology,The First Affiliated Hospital of Harbin Medical University, Harbin 150007, China
| | - Jihong Wang
- Department of oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Xiaojing Chen
- Department of oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yuhang Zhou
- Department of oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Kai You
- Department of Chest Surgery,The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Han Li
- Department of oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yi Jiang
- Department of oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Zhiyao Zhang
- Department of oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Xinrui Du
- Department of oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yunwei Han
- Department of oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
7
|
Lee J, Roh JL. Selenium and selenoproteins: key regulators of ferroptosis and therapeutic targets in cancer. J Mol Med (Berl) 2025:10.1007/s00109-025-02563-8. [PMID: 40527925 DOI: 10.1007/s00109-025-02563-8] [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: 03/14/2025] [Revised: 06/01/2025] [Accepted: 06/03/2025] [Indexed: 06/20/2025]
Abstract
The interplay between selenium (Se) metabolism and ferroptosis presents a compelling area of study in cancer biology. This review synthesizes the current understanding of key pathways implicated in ferroptosis susceptibility, with a focus on the role of selenoproteins, particularly glutathione peroxidase 4 (GPX4), which mitigates lipid peroxidation and prevents ferroptotic cell death through the system Xc-/GSH axis. Additionally, selenoprotein P contributes to Se transport, playing a crucial role in ferroptosis resistance observed in certain cancers. Targeting Se pathways, especially disrupting GPX4 and selenoprotein P functions, offers promising avenues for cancer therapy. The differential dependence of cancer cells on Se and selenoproteins highlights the potential for selective induction of ferroptosis in malignant cells. Future research should focus on unraveling the mechanistic underpinnings of Se-mediated ferroptosis and exploring combinatorial therapeutic strategies. This review sets the stage for innovative approaches that leverage Se metabolism to enhance cancer treatment efficacy through ferroptosis modulation. KEY MESSAGES: Selenium (Se) and selenoproteins regulate ferroptosis, a lipid peroxidation-driven form of cell death. GPX4, a Se-dependent enzyme, defends cells by neutralizing lipid hydroperoxides. Xc-/GSH/GPX4 and FSP1-CoQ10 pathways are critical in modulating ferroptosis susceptibility. Selenoprotein P, SEPHS2, and SQOR highlight vulnerabilities in Se-dependent cancer cell survival. Se's role in balancing antioxidant defense and ferroptosis offers therapeutic insights.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea
- Logsynk, Seoul, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do, 13496, Republic of Korea.
- Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea.
| |
Collapse
|
8
|
Li GZ, Liu JY, Zhou H. Ferroptosis: A novel therapeutic target for diabetic cardiomyopathy. World J Diabetes 2025; 16:104665. [DOI: 10.4239/wjd.v16.i6.104665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/24/2025] [Accepted: 05/07/2025] [Indexed: 06/13/2025] Open
Abstract
Ferroptosis is a new type of programmed cell death caused by the accumulation of iron-dependent lipid peroxides, and it plays a role in the occurrence and progression of diverse diseases. Diabetic cardiomyopathy (DCM), a serious cardiovascular complication in patients with diabetes, eventually progresses to refractory heart failure (HF), which increases the risk of hospitalization for HF and cardiovascular death in patients with diabetes. Despite glycemic control, effective strategies to prevent DCM onset are currently lacking. Accumulating evidence suggests that ferroptosis is involved in oxidative stress, inflammation, and abnormal autophagy in diabetic myocardium, which plays an important role in myocardial apoptosis, hypertrophy, and cardiac fibrosis. The inhibition of ferroptosis can relieve DCM. Presently, ferroptosis inhibitors have been broadly suggested for the treatment of iron overload-related cardiomyopathy. This article reviewed relevant studies to offer a new therapeutic target for DCM.
Collapse
Affiliation(s)
- Gui-Zhi Li
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Jia-Yin Liu
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
| | - Hong Zhou
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
9
|
Chung JY, Knutson BA. Bypassing the guardian: regulated cell death pathways in p53-mutant cancers. Cell Mol Biol Lett 2025; 30:68. [PMID: 40517236 PMCID: PMC12166615 DOI: 10.1186/s11658-025-00751-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Approximately half of all cancers bear mutations in the tumor suppressor p53. Despite decades of research studying p53 function, treatment of p53-mutant cancers remains challenging owing to the effects of p53 mutations on many complex and interrelated signaling networks that promote tumor metastasis and chemoresistance. Mutations in p53 promote tumor survival by dysregulating cellular homeostasis and preventing activation of regulated cell death (RCD) pathways, which normally promote organismal health by eliminating dysregulated cells. Activation of RCD is a hallmark of effective cancer therapies, and p53-mutant cancers may be particularly susceptible to activation of certain RCD pathways. In this review, we discuss four RCD pathways that are the targets of emerging cancer therapeutics to treat p53-mutant cancers. These RCD pathways include E2F1-dependent apoptosis, necroptosis, mitochondrial permeability transition-driven necrosis, and ferroptosis. We discuss mechanisms of RCD activation, effects of p53 mutation on RCD activation, and current pharmaceutical strategies for RCD activation in p53-mutant cancers.
Collapse
Affiliation(s)
- Jonathan Y Chung
- Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Bruce A Knutson
- Department of Biochemistry and Molecular Biology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
10
|
Yang J, Wang Y, Liu F, Zhang Y, Han F. Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review). Int J Mol Med 2025; 55:97. [PMID: 40314096 PMCID: PMC12045474 DOI: 10.3892/ijmm.2025.5538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025] Open
Abstract
Ferroptosis is a unique mode of cell death driven by iron‑dependent phospholipid peroxidation, and its mechanism primarily involves disturbances in iron metabolism, imbalances in the lipid antioxidant system and accumulation of lipid peroxides. Protein processing, modification and folding in the endoplasmic reticulum (ER) are closely related regulatory processes that determine cell function, fate and survival. The uncontrolled proliferative capacity of malignant cells generates an unfavorable microenvironment characterized by high metabolic demand, hypoxia, nutrient deprivation and acidosis, which promotes the accumulation of misfolded or unfolded proteins in the ER, leading to ER stress (ERS). Ferroptosis and ERS share common pathways in several diseases, and the two interact to affect cell survival and death. Additionally, cell death pathways are not linear signaling cascades, and different pathways of cell death may be interrelated at multiple levels. Ferroptosis and ERS in ovarian cancer (OC) have attracted increasing research interest; however, both are discussed separately regarding OC. The present review aims to summarize the associations and potential links between ferroptosis and ERS, aiming to provide research references for the development of therapeutic approaches for the management of OC.
Collapse
Affiliation(s)
- Jiaqi Yang
- Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yu Wang
- Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Fangyuan Liu
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yizhong Zhang
- Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Fengjuan Han
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
11
|
Alarcón-Veleiro C, López-Calvo I, Berjawi L, Lucio-Gallego S, Mato-Basalo R, Quindos-Varela M, Lesta-Mellid R, Santamarina-Caínzos I, Varela-Rodríguez S, Fraga M, Quintela M, Vizoso-Vázquez A, Arufe MC, Fafián-Labora J. Ferroptosis: An emerging strategy for managing epithelial ovarian cancer. Biomed Pharmacother 2025; 187:118065. [PMID: 40306179 DOI: 10.1016/j.biopha.2025.118065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/30/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
Ferroptosis is a regulated form of cell death characterised by iron-dependent lipid peroxidation, a process intricately linked to cellular redox homeostasis. This form of cell death is induced by the accumulation of intracellular iron and the subsequent generation of reactive oxygen species (ROS), which leads to lipid peroxidation and ultimately cell death. Ferroptosis is distinct from traditional forms of cell death, such as apoptosis, and holds significant therapeutic potential, particularly in cancers harboring rat sarcoma virus (RAS) mutations, such as epithelial ovarian cancer (EOC). EOC is notoriously resistant to conventional therapies and is associated with a poor prognosis. In this review, we examine recent progress in the understanding of ferroptosis, with a particular focus on its redox biology and the complex regulatory networks involved. We also propose a novel classification system for ferroptosis modulators, grouping them into six categories (I, II, III, IV, V and VI) based on their mechanisms of action and their roles in modulating cellular redox status. By refining these categories, we aim to provide deeper insights into the role of ferroptosis in cancer biology, especially in EOC, and to identify potential therapeutic avenues. We propose that further investigation of ferroptosis in the context of redox biology could reveal novel biomarkers and therapeutic targets, offering promising strategies to overcome resistance mechanisms and improve clinical outcomes for patients with EOC and other treatment-resistant cancers.
Collapse
Affiliation(s)
- C Alarcón-Veleiro
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - I López-Calvo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain; Centro Interdisciplinar de Química de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain; Instituto de Investigación Biomédica de A Coruña (INIBIC), Rúa as Xubias 84, A Coruña 15006, Spain
| | - L Berjawi
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - S Lucio-Gallego
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - R Mato-Basalo
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain
| | - M Quindos-Varela
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - R Lesta-Mellid
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - I Santamarina-Caínzos
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - S Varela-Rodríguez
- Translational Cancer Research Group, A Coruña Biomedical Research Institute (INIBIC), Carretera del Pasaje s/n, A Coruña 15006, UK; Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - M Fraga
- Department of Anatomical Pathology, University Hospital Complex A Coruña, As Xubias 84, A Coruña 15006, Spain
| | - M Quintela
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff CF24 4HQ, UK
| | - A Vizoso-Vázquez
- Grupo EXPRELA, Departamento de Bioloxía, Facultade de Ciencias, Rúa da Fraga, A Coruña 15071, Spain
| | - M C Arufe
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| | - J Fafián-Labora
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, INIBIC-Complejo Hospitalario Universitario A Coruña (CHUAC), Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña (UDC), A Coruña 15008, Spain.
| |
Collapse
|
12
|
Li M, Li Y, Yao X, Liu Y, Cai K, Yang H, Luo Z. Optically controllable nanoregulators enable tumor-specific pro-ferroptosis lipometabolic reprogramming for in-situ adjuvant-free vaccination. Biomaterials 2025; 317:123096. [PMID: 39805186 DOI: 10.1016/j.biomaterials.2025.123096] [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: 10/29/2024] [Revised: 12/26/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
In-situ tumor vaccination remains challenging due to difficulties in the exposure and presentation of tumor-associated neoantigens (TANs). In view of the central role of lipid metabolism in cell fate determination and tumor-immune cell communication, here we report a photo-controlled lipid metabolism nanoregulator (PLMN) to achieve robust in-situ adjuvant-free vaccination, which is constructed through hierarchically integrating photothermal-inducible arachidonate 15-lipoxygenase (ALOX15)-expressing plasmids, cypate and FIN56 into cationic liposomes. Near-infrared light (NIR) stimulation triggers on-demand ALOX15 editing and causes excessive accumulation of downstream pro-ferroptosis lipid metabolites. PLMN treatment enables efficient TAN release through ferroptosis-dependent membrane perturbation and facilitates their capture and processing by antigen-presenting cells via cationic lipid-mediated TAN enrichment. Meanwhile, upregulation of ALOX15-associated lipid metabolites also enhances M2-to-M1 phenotypic transition of tumor-associated macrophages through regulating tumor-macrophage metabolic crosstalk. PLMN treatment significantly enhance the robustness and durability of adaptive antitumor immunity in vivo, offering an approach for in-situ tumor vaccination in the clinic.
Collapse
Affiliation(s)
- Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Yanan Li
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Xuemei Yao
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Yingqi Liu
- School of Life Science, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing, 400044, China
| | - Huocheng Yang
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
13
|
Dai X, Zheng Y, Cui J, Zeng Y, Yang B, Zhang Z. Nanodrug delivery systems targeting ferroptosis as an innovative therapeutic approach for Rheumatoid Arthritis. Mater Today Bio 2025; 32:101804. [PMID: 40343168 PMCID: PMC12059336 DOI: 10.1016/j.mtbio.2025.101804] [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: 02/13/2025] [Revised: 04/02/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
Rheumatoid Arthritis (RA) is a chronic inflammatory disease characterized by joint inflammation, progressive cartilage degradation, and bone erosion. Recent research has implicated ferroptosis not only in autoimmune hepatitis but also in the pathogenesis and progression of autoimmune disorders like RA. Consequently, numerous therapeutic strategies have begun to target the ferroptosis pathway, particularly in the design and development of nanodrug delivery systems (NDDSs). While previous reviews have comprehensively discussed the mechanisms of ferroptosis, related signaling pathways, and NDDS materials, recent studies have further elucidated the interplay between ferroptosis and various metabolic pathways, providing a robust theoretical basis for the design of NDDS-based ferroptosis strategies. This review focuses on investigating the role of ferroptosis in the development of RA, aiming to elucidate how targeting ferroptosis can offer novel therapeutic concepts and potential treatments for RA patients. Specifically, it summarizes the design strategies of ferroptosis-based NDDSs via different pathways and highlights the feasibility of RA treatment regimens based on the ferroptosis mechanism. Furthermore, the review critically discusses the current limitations of NDDSs and offers perspectives on future research directions in this field.
Collapse
Affiliation(s)
- Xiaolin Dai
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Yu Zheng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, PR China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, 646000, PR China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, PR China
- Sichuan-Chongqing Joint Key Laboratory of Metabolic Vascular Diseases, Luzhou, 646000, PR China
| | - Jianrong Cui
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Yuqi Zeng
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Bo Yang
- Department of Pharmacy, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Zhanlin Zhang
- Irradiation Preservation and Effect Key Laboratory of Sichuan Province, School of Bioscience and Technology, Chengdu Medical College, Chengdu, 610500, PR China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, PR China
| |
Collapse
|
14
|
Zhang Y, Shi H, Wang Y, Liu W, Li G, Li D, Wu W, Wu Y, Zhang Z, Ji Y, Zhu C, Bai W, Lei H, Xu H, Zhong H, Han B, Yang L, Liu L, Wang W, Zhao Y, Zhang Y, Wu Y. Noscapine derivative 428 suppresses ferroptosis through targeting GPX4. Redox Biol 2025; 83:103635. [PMID: 40305884 DOI: 10.1016/j.redox.2025.103635] [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: 02/01/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Inhibiting ferroptosis represents a promising strategy to combat ferroptosis-related diseases. Here we show that 428, a selenide-containing noscapine derivative, effectively inhibits ferroptosis in various cell lines by enhancing the stability and activity of GPX4. TRIM41 was identified as a novel E3 ubiquitin ligase of GPX4 and 428 was demonstrated to bind to the selenocysteine residue Sec46 of GPX4 via the formation of a transient and reversible Se-Se bond, thereby blocking the interaction between GPX4 and TRIM41, stabilizing GPX4 and enhancing its activity. This unique dynamic covalent binding mode was preliminarily validated by structure-activity relationship analysis and molecular docking studies. Importantly, we demonstrated that 428 treatment alleviates bleomycin-induced pulmonary fibrosis in vivo by inhibiting ferroptosis. Overall, our studies identified a novel stabilizer and activator of GPX4, offering a potential therapeutic approach for the treatment of ferroptosis-related diseases and uncovering a new mechanism for regulating GPX4 degradation.
Collapse
Affiliation(s)
- Youping Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haoliang Shi
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yingying Wang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wanting Liu
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guanyi Li
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Defeng Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenxuan Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yunzhao Wu
- Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW, Cambridge, UK
| | - Ziwei Zhang
- Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW, Cambridge, UK
| | - Yanjie Ji
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chujiao Zhu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenhui Bai
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hu Lei
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hanzhang Xu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hua Zhong
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Baohui Han
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ligen Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Wang
- Department of Pharmacology and Toxicology and BIO5 Institute, University of Arizona, USA
| | - Yaxue Zhao
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| | - Yongqiang Zhang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Yingli Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
15
|
Ruan Y, Zhang L, Zhang L, Zhu K. Therapeutic Approaches Targeting Ferroptosis in Cardiomyopathy. Cardiovasc Drugs Ther 2025; 39:595-613. [PMID: 37930587 DOI: 10.1007/s10557-023-07514-4] [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] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
The term cardiomyopathy refers to a group of heart diseases that cause severe heart failure over time. Cardiomyopathies have been proven to be associated with ferroptosis, a non-apoptotic form of cell death. It has been shown that some small molecule drugs and active ingredients of herbal medicine can regulate ferroptosis, thereby alleviating the development of cardiomyopathy. This article reviews recent discoveries about ferroptosis, its role in the pathogenesis of cardiomyopathy, and the therapeutic options for treating ferroptosis-associated cardiomyopathy. The article aims to provide insights into the basic mechanisms of ferroptosis and its treatment to prevent cardiomyopathy and related diseases.
Collapse
Affiliation(s)
- Yanqian Ruan
- School of Public Health, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center of Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Ling Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Lina Zhang
- School of Public Health, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center of Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
| | - Keyang Zhu
- School of Public Health, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center of Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China.
| |
Collapse
|
16
|
Ge T, Wang Y, Han Y, Bao X, Lu C. Exploring the Updated Roles of Ferroptosis in Liver Diseases: Mechanisms, Regulators, and Therapeutic Implications. Cell Biochem Biophys 2025; 83:1445-1464. [PMID: 39543068 DOI: 10.1007/s12013-024-01611-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
Ferroptosis, a newly discovered mode of cell death, is a type of iron-dependent regulated cell death characterized by intracellular excessive lipid peroxidation and imbalanced redox. As the liver is susceptible to oxidative damage and the abnormal iron accumulation is a major feature of most liver diseases, studies on ferroptosis in the field of liver diseases are of great interest. Studies show that targeting the key regulators of ferroptosis can effectively alleviate or even reverse the deterioration process of liver diseases. System Xc- and glutathione peroxidase 4 are the main defense regulators of ferroptosis, while acyl-CoA synthetase long chain family member 4 is a key enzyme causing peroxidation in ferroptosis. Generally speaking, ferroptosis should be suppressed in alcoholic liver disease, non-alcoholic fatty liver disease, and drug-induced liver injury, while it should be induced in liver fibrosis and hepatocellular carcinoma. In this review, we summarize the main regulators involved in ferroptosis and then the mechanisms of ferroptosis in different liver diseases. Treatment options of drugs targeting ferroptosis are further concluded. Determining different triggers of ferroptosis can clarify the mechanism of ferroptosis occurs at both physiological and pathological levels.
Collapse
Affiliation(s)
- Ting Ge
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yang Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Yiwen Han
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
17
|
Leak L, Wang Z, Joseph AJ, Johnson B, Chan AA, Decosto CM, Magtanong L, Ko PJ, Lee WC, Ritho J, Manukian S, Millner A, Chitkara S, Salinas JJ, Skouta R, Rees MG, Ronan MM, Roth JA, Myers CL, Moffat J, Boone C, Bensinger SJ, Nathanson DA, Atilla-Gokcumen GE, Moding EJ, Dixon SJ. Tegavivint triggers TECR-dependent nonapoptotic cancer cell death. Nat Chem Biol 2025:10.1038/s41589-025-01913-4. [PMID: 40419770 DOI: 10.1038/s41589-025-01913-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/16/2025] [Indexed: 05/28/2025]
Abstract
Small molecules that induce nonapoptotic cell death are of fundamental mechanistic interest and may be useful to treat certain cancers. Here we report that tegavivint, a drug candidate undergoing human clinical trials, can activate a unique mechanism of nonapoptotic cell death in sarcomas and other cancer cells. This lethal mechanism is distinct from ferroptosis, necroptosis and pyroptosis and requires the lipid metabolic enzyme trans-2,3-enoyl-CoA reductase (TECR). TECR is canonically involved in the synthesis of very-long-chain fatty acids but appears to promote nonapoptotic cell death in response to CIL56 and tegavivint via the synthesis of the saturated long-chain fatty acid palmitate. These findings outline a lipid-dependent nonapoptotic cell death mechanism that can be induced by a drug candidate currently being tested in humans.
Collapse
Affiliation(s)
- Logan Leak
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Ziwei Wang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alby J Joseph
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Brianna Johnson
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Alyssa A Chan
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | | | - Pin-Joe Ko
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Joan Ritho
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Sophia Manukian
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Alec Millner
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Shweta Chitkara
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Jennifer J Salinas
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rachid Skouta
- Department of Chemistry, University of Massachusetts, Amherst, Amherst, MA, USA
- Department of Biology, University of Massachusetts, Amherst, Amherst, MA, USA
| | | | | | | | - Chad L Myers
- Department of Computer Science and Engineering, Bioinformatics and Computational Biology Graduate Program, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Jason Moffat
- Program in Genetics & Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Charles Boone
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- RIKEN Center for Sustainable Resource Science, Saitama, Japan
| | - Steven J Bensinger
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- UCLA Lipidomics Laboratory, University of California, Los Angeles, Los Angeles, CA, USA
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - G Ekin Atilla-Gokcumen
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
18
|
Xu H, Li J, Peng S, Bai Y, Tang W. From exposure to innovation: decoding aromatic amines' role in bladder cancer mechanisms. Discov Oncol 2025; 16:888. [PMID: 40410554 PMCID: PMC12102015 DOI: 10.1007/s12672-025-02730-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 05/16/2025] [Indexed: 05/25/2025] Open
Abstract
OBJECTIVE Aromatic amines (AAs) have been verified as a risk factor for bladder cancer (BCa). Most existing studies have focused on specific AAs types in BCa and assessed the impact of exposure to AAs on the prognosis of BCa patients; however, there is no comprehensive exploration of the mechanism of action. Therefore, this study explored the core hub genes (CHGs) involved in the interaction between major AAs and BCa through multi-database joint analysis to clarify the molecular mechanism of the AAS-induced occurrence and development of BCa and provide innovative insights in the diagnosis and treatment of AAS-induced BCa. METHODS After the toxicity analysis of AAs, the toxicity regulatory network of AAs in BCa was constructed through network toxicology, and the targets that showed the causal relationship with BCa were screened by Mendelian randomization (MR) analysis. Comprehensive mechanism exploration, molecular docking and drug prediction analysis were conducted on CHGs defined by the protein-protein interaction (PPI) network. RESULTS The seven CHGs for the five AAs with different degrees of carcinogenicity to exert toxicity to BCa regulated the occurrence and development of BCa via multiple signaling pathways. Molecular docking confirmed the potential of the activation of these pathways caused by AAs. The results of drug prediction analysis suggested that rapamycin had a potential therapeutic prospect for AAs-induced BCa. CONCLUSION This study reveals the underlying molecular mechanism by which exposure to AAs leads to the occurrence and development of BCa, providing novel preventive and therapeutic insights for populations exposed to this exposure factor.
Collapse
Affiliation(s)
- Haoyu Xu
- Department of Urology, First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjia Gang, Yuzhong District, Chongqing, 400016, China
| | - Junwu Li
- Department of Urology, First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjia Gang, Yuzhong District, Chongqing, 400016, China
| | - Senlin Peng
- Department of Urology, First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjia Gang, Yuzhong District, Chongqing, 400016, China
| | - Yuanyuan Bai
- Department of Urology, First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjia Gang, Yuzhong District, Chongqing, 400016, China
| | - Wei Tang
- Department of Urology, First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuanjia Gang, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
19
|
Li Q, Cheng Y, Yang C, Tian M, Wang X, Li D, Li X, Qu J, Zhou S, Zheng L, Tong Q. Targeting the Exonic Circular OGT RNA/O-GlcNAc Transferase/Forkhead Box C1 Axis Inhibits Asparagine- and Alanine-Mediated Ferroptosis Repression in Neuroblastoma Progression. RESEARCH (WASHINGTON, D.C.) 2025; 8:0703. [PMID: 40416363 PMCID: PMC12099056 DOI: 10.34133/research.0703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/27/2025]
Abstract
The disruption of ferroptosis, an emerging form of programmed cell death, is crucial in the development and aggressiveness of tumors. Meanwhile, the mechanisms and treatments that control ferroptosis in neuroblastoma (NB), a prevalent extracranial cancer in children, are still unknown. In this study, forkhead box C1 (FOXC1) and O-GlcNAc transferase (OGT) are identified as regulators of asparagine- and alanine-mediated ferroptosis repression in NB. Mechanistically, OGT facilitates FOXC1 stabilization via inducing O-GlcNAcylation in liquid condensates to increase the expression of asparagine synthetase (ASNS) and glutamate pyruvate transaminase 2 (GPT2), resulting in asparagine and alanine biogenesis, and subsequent synthesis of cystathionine β-synthase (CBS) or ferritin heavy chain 1 (FTH1). Meanwhile, exonic circular OGT RNA (ecircOGT) is able to encode a novel protein (OGT-570aa) containing domain essential for binding of OGT to FOXC1, which competitively decreases the OGT-FOXC1 interaction. Preclinically, miconazole nitrate facilitates the interaction of OGT-570aa with FOXC1, suppresses ferroptosis resistance of NB cells, and inhibits their growth, invasion, and metastasis. In clinical NB cases, higher OGT, FOXC1, ASNS, GPT2, CBS, or FTH1 levels are correlated with worse survival, while lower ecircOGT or OGT-570aa expression is associated with tumor progression. These results indicate that targeting the ecircOGT/OGT/FOXC1 axis inhibits asparagine- and alanine-mediated ferroptosis repression in NB progression.
Collapse
Affiliation(s)
- Qilan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Yang Cheng
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Chunhui Yang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Minxiu Tian
- Department of Pathology, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Xiaojing Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
- Department of Geriatrics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Dan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Xinyue Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Jiaying Qu
- Department of Pathology, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Shunchen Zhou
- Department of Pathology, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, P. R. China
| |
Collapse
|
20
|
Li B, Hai E, Song Y, Zhang J. Autophagic Degradation of GPX4 Mediates Ferroptosis During Sheep Sperm Cryopreservation. Vet Sci 2025; 12:490. [PMID: 40431583 PMCID: PMC12115632 DOI: 10.3390/vetsci12050490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Ferroptosis is implicated in cryodamage to sheep sperm, potentially due to glutathione peroxidase 4 (GPX4) degradation during freezing; however, the pathway underlying GPX4 degradation remains unclear. In this study, a comparison of cryoprotective effects between the autophagy inhibitor chloroquine (CQ) and the ubiquitination inhibitor MG132 revealed that 5 μM CQ treatment significantly enhanced the motility (p < 0.01) and sperm plasma membrane integrity rate (p < 0.01) of frozen-thawed sperm; no protective effects were observed in any MG132 treatment group. Mechanistic analysis indicated that CQ treatment substantially restored GPX4 protein expression (p < 0.01), and concurrently reduced lipid peroxidation (p < 0.01) and free iron ion accumulation (p < 0.01), in frozen-thawed sperm. These findings suggest that GPX4 degradation during cryopreservation occurs via the autophagy pathway. This study established a ferroptosis-GPX4-autophagy axis during sheep sperm cryopreservation and identified autophagy-mediated GPX4 loss as a potential target for enhancing sperm cryoprotection.
Collapse
Affiliation(s)
| | | | | | - Jiaxin Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (B.L.); (E.H.); (Y.S.)
| |
Collapse
|
21
|
Zheng J, Zhang W, Ito J, Henkelmann B, Xu C, Mishima E, Conrad M. N-acetyl-l-cysteine averts ferroptosis by fostering glutathione peroxidase 4. Cell Chem Biol 2025; 32:767-775.e5. [PMID: 40311609 DOI: 10.1016/j.chembiol.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/08/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
N-acetyl-l-cysteine (NAC) is a medication and a widely used antioxidant in cell death research. Despite its somewhat obscure mechanism of action, its role in inhibiting ferroptosis is gaining increasing recognition. In this study, we demonstrate that NAC treatment rapidly replenishes the intracellular cysteine pool, reinforcing its function as a prodrug for cysteine. Interestingly, its enantiomer, N-acetyl-d-cysteine (d-NAC), which cannot be converted into cysteine, also exhibits a strong anti-ferroptotic effect. We further clarify that NAC, d-NAC, and cysteine all act as direct reducing substrates for GPX4, counteracting lipid peroxidation. Consequently, only GPX4-rather than system xc-, glutathione biosynthesis, or ferroptosis suppressor protein 1-is necessary for NAC and d-NAC to prevent ferroptosis. Additionally, we identify a broad range of reducing substrates for GPX4 in vitro, including β-mercaptoethanol. These findings provide new insights into the mechanisms underlying the protective effects of NAC and other potential GPX4-reducing substrates against ferroptosis.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Weijia Zhang
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Junya Ito
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Laboratory of Food Function Analysis, Tohoku University Graduate School of Agricultural Science, Sendai, Miyagi 980-8572, Japan
| | - Bernhard Henkelmann
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Chenxi Xu
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Eikan Mishima
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8574, Japan
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, 85748 Garching, Germany.
| |
Collapse
|
22
|
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; 109:102770. [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
|
23
|
Chen J, Fu Y, Weng S, He J, Dong C. Vitamin C Inhibits Scale Drop Disease Virus Infectivity by Targeting Nrf2 to Reduce Ferroptosis. Antioxidants (Basel) 2025; 14:576. [PMID: 40427458 PMCID: PMC12108497 DOI: 10.3390/antiox14050576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/04/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Scale drop disease virus (SDDV) poses an escalating threat to global aquaculture, prompting an urgent need for research. Our study found that SDDV infection upregulates genes related to iron, oxidative stress, and lipid metabolism, causing iron overload, reactive oxygen species (ROS) accumulation, and ultimately ferroptosis. Among the tested antioxidants, vitamin C (VC) demonstrated the most potent inhibitory effect in mandarin fish, reducing SDDV-induced mortality by 37.5%. qPCR and IFA results showed that VC effectively suppressed SDDV infection; decreased ROS, lipid peroxidation (LPO), and iron levels; and enhanced glutathione peroxidase 4 (GPX4) expression in infected cells. Mechanistically, VC's inhibitory effect was reversed by the nuclear factor erythroid 2-related factor 2 (Nrf2) inhibitor ML-385, indicating an Nrf2-dependent pathway. VC promoted Nrf2 nuclear translocation and activated downstream antioxidant genes. Moreover, VC modulated inflammation by regulating pro- and anti-inflammatory factors. These findings suggest VC as a promising therapeutic for SDDV infection.
Collapse
Affiliation(s)
- Jiaming Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
| | - Yuting Fu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Jianguo He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Chuanfu Dong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, China; (J.C.); (Y.F.); (S.W.); (J.H.)
- Institute of Aquatic Economic Animals, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou 510275, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| |
Collapse
|
24
|
Hao X, Wang Y, Hou MJ, Yang YX, Liao L, Chen T, Wang P, Chen X, Zhu BT. Strong protection by bazedoxifene against chemically-induced ferroptotic neuronal death in vitro and in vivo. Cell Commun Signal 2025; 23:218. [PMID: 40336106 PMCID: PMC12060420 DOI: 10.1186/s12964-025-02209-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
Ferroptosis, a form of regulated cell death associated with glutathione depletion and excess lipid peroxidation, can be induced in cultured cells by chemicals (e.g., erastin and RSL3). It has been shown that protein disulfide isomerase (PDI) is a mediator of chemically-induced ferroptosis and also a crucial target for ferroptosis protection. The present study reports that bazedoxifene (BAZ), a selective estrogen receptor modulator, is an inhibitor of PDI and can strongly rescue neuronal cells from chemically-induced oxidative ferroptosis. We find that BAZ can directly bind to PDI and inhibit its catalytic activity. Computational modeling analysis reveals that BAZ forms a hydrogen bond with PDI's His256 residue. Inhibition of PDI by BAZ markedly reduces iNOS and nNOS dimerization (i.e., catalytic activation) and NO accumulation, and these effects of BAZ are associated with reductions in cellular ROS and lipid-ROS and protection against chemically-induced ferroptosis. In addition, the direct antioxidant activity of BAZ may also partially contribute to its protection against chemically-induced ferroptosis. In vivo animal experiments show that mice treated with BAZ are strongly protected against kainic acid-induced oxidative hippocampal neuronal injury and memory deficits. Together, these results reveal that BAZ is a potent inhibitor of PDI and can strongly protect against chemically-induced ferroptosis in hippocampal neurons both in vitro and in vivo. This work provides evidence for an estrogen receptor-independent, PDI-mediated novel mechanism of neuroprotection by BAZ.
Collapse
Affiliation(s)
- Xiangyu Hao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Yifan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Ming-Jie Hou
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Yong Xiao Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Lixi Liao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Tongxiang Chen
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Xiaojun Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
25
|
Xu M, Xu B. Protein lipidation in the tumor microenvironment: enzymology, signaling pathways, and therapeutics. Mol Cancer 2025; 24:138. [PMID: 40335986 PMCID: PMC12057185 DOI: 10.1186/s12943-025-02309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/18/2025] [Indexed: 05/09/2025] Open
Abstract
Protein lipidation is a pivotal post-translational modification that increases protein hydrophobicity and influences their function, localization, and interaction network. Emerging evidence has shown significant roles of lipidation in the tumor microenvironment (TME). However, a comprehensive review of this topic is lacking. In this review, we present an integrated and in-depth literature review of protein lipidation in the context of the TME. Specifically, we focus on three major lipidation modifications: S-prenylation, S-palmitoylation, and N-myristoylation. We emphasize how these modifications affect oncogenic signaling pathways and the complex interplay between tumor cells and the surrounding stromal and immune cells. Furthermore, we explore the therapeutic potential of targeting lipidation mechanisms in cancer treatment and discuss prospects for developing novel anticancer strategies that disrupt lipidation-dependent signaling pathways. By bridging protein lipidation with the dynamics of the TME, our review provides novel insights into the complex relationship between them that drives tumor initiation and progression.
Collapse
Affiliation(s)
- Mengke Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China.
| |
Collapse
|
26
|
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
|
27
|
Dangi K, Kumar V, Mittal D, Yadav P, Malik M, Verma AK. Nanotherapeutics induced redox resetting of oxidative and nitrosative stress: targeting glutathione-depletion in cancer. Nanomedicine (Lond) 2025; 20:955-965. [PMID: 40192277 PMCID: PMC12051575 DOI: 10.1080/17435889.2025.2489918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
Cancer cells display a distinctive defense mechanism against any exogenous moieties that renders all treatments inefficient. Glutathione, a thiol tripeptide plays a paradoxical role in cancer as intracellular glutathione (GSH) are voracious scavengers of free radicals produced by chemotherapy, generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS). Cancer cells show Warburg effect, wherein the intracellular GSH levels are exceptionally enhanced to overcome the oxidative stress created by ROS/RNS production or by the other free radicals generated as side products of intracellular redox reactions. Therefore, redox resetting is essential to maintain the redox homeostasis for cell survival and their proliferation and trigger escalation of GSH levels. Nanotherapeutics have facilitated the targeted delivery of GSH-depleting agents in combination with radiotherapy, chemotherapy, and novel therapeutic interventions including chemodynamic therapy (CDT), photodynamic therapy (PDT), ferroptosis induction, sonodynamic therapy (SDT), and immunotherapy are being explored. This review aims to compile the strategic role of GSH in cancer cells, the importance of nanotherapeutics for GSH depletion in cancer to target numerous forms of programmed cell death (PCD), including apoptosis, ferroptosis, necroptosis, and autophagy.
Collapse
Affiliation(s)
- Kapil Dangi
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi, India
- Department of Zoology, Deshbandhu College, University of Delhi, Delhi, India
| | - Vijay Kumar
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi, India
- Department of Anatomy, School of Medicine, University of Galway, Galway, Ireland
| | - Disha Mittal
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi, India
- Department of Life Sciences, School of Biosciences and Technology, Galgotias University, Greater Noida, India
| | - Pooja Yadav
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi, India
| | - Mansi Malik
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi, India
| | - Anita Kamra Verma
- Nano-Biotech Lab, Kirori Mal College, University of Delhi, Delhi, India
- Delhi School of Public Health, Institution of Eminence, University of Delhi, Delhi, India
| |
Collapse
|
28
|
Loftus LV, Rolle LTA, Wang B, Pienta KJ, Amend SR. Dysregulation of Labile Iron Predisposes Chemotherapy Resistant Cancer Cells to Ferroptosis. Int J Mol Sci 2025; 26:4193. [PMID: 40362430 PMCID: PMC12072162 DOI: 10.3390/ijms26094193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Despite centuries of research, metastatic cancer remains incurable due to resistance to all conventional cancer therapeutics. Alternative strategies leveraging non-proliferative vulnerabilities in cancer are required to overcome cancer recurrence. Ferroptosis is an iron dependent cell death pathway that has shown promising pre-clinical activity in several contexts of therapeutic resistant cancer. However, ferroptosis sensitivity is highly variable across tissue types and cell states, posing a challenge for clinical translation. We describe a convergent phenotype induced by chemotherapy where cells surviving chemotherapy have dysregulated iron homeostasis, regardless of initial cell type or chemotherapy used. Elevated labile iron levels are counteracted by NRF2 signaling, yet the resulting antioxidant programs do not alleviate the labile iron burden. Selectively inhibiting GPX4 leads to uniform susceptibility to ferroptosis in surviving cells, highlighting the common reliance on lipid peroxidation defenses. Cellular iron dysregulation is a vulnerability of chemoresistant cancer cells that can be leveraged by triggering ferroptosis.
Collapse
Affiliation(s)
- Luke V. Loftus
- Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Cancer Ecology Center, Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Louis T. A. Rolle
- Cancer Ecology Center, Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Bowen Wang
- Cancer Ecology Center, Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Pathobiology Program, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Kenneth J. Pienta
- Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Cancer Ecology Center, Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Pathobiology Program, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Sarah R. Amend
- Cellular and Molecular Medicine Program, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Cancer Ecology Center, Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Pathobiology Program, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
29
|
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
|
30
|
Direksunthorn T, T Ahmed A, Pluetrattanabha N, Uthirapathy S, Ballal S, Singh A, Al-Hetty HRAK, Devi A, Sharma GC, Yumashev A. Ferroptosis in immune chaos: Unraveling its impact on disease and therapeutic potential. J Physiol Biochem 2025:10.1007/s13105-025-01078-7. [PMID: 40237936 DOI: 10.1007/s13105-025-01078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
Since its introduction in 2012, ferroptosis has garnered significant attention from researchers over the past decade. Unlike autophagy and apoptosis, ferroptosis is an atypical iron-dependent programmed cell death that falls under necrosis. It is regulated by various cellular metabolic and signaling processes, which encompass amino acid, lipid, iron, and mitochondrial metabolism. The initiation of ferroptosis occurs through iron-dependent phospholipid peroxidation. Notably, ferroptosis exhibits a dual effect and is associated with various diseases. A significant challenge lies in managing autoimmune disorders with unknown origins that stem from the reactivation of the immune system. Two contributing factors to autoimmunity are the aberrant stimulation of cell death and the inadequate clearance of dead cells, which can expose or release intracellular components that activate the immune response. Ferroptosis is distinct from other forms of cell death, such as apoptosis, necroptosis, autophagy, and pyroptosis, due to its unique morphological, biochemical, and genetic characteristics and specific relationship with cellular iron levels. Recent studies indicate that immune cells can both induce and undergo ferroptosis. To better understand how ferroptosis influences immune responses and its imbalance in disease, a molecular understanding of the relationship between ferroptosis and immunity is essential. Consequently, further research is needed to develop immunotherapeutics that target ferroptosis. This review primarily focuses on the role of ferroptosis in immune-related disorders.
Collapse
Affiliation(s)
| | | | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | | | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
31
|
Wu Z, Zhang G, Shang Y, Huang J, Liu Y, Zhou H, Wang T. New curcumin derivative induces ferroptosis in MCF-7 cells through activating SLC7A11/GPX4 axis. Bioorg Med Chem 2025; 121:118078. [PMID: 39965986 DOI: 10.1016/j.bmc.2025.118078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/28/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025]
Abstract
Previous experiments have revealed that curcumin exerts potential antitumor effect by inducing apoptosis and ferroptosis of tumor cells. However, its low solubility and bioavailability, as well as fast metabolism limit its clinical use. The structural modification of curcumin is beneficial for the discovery of potential candidate drugs for cancer treatment. Here, three new series of curcumin derivatives including 25 compounds were synthesized at active sites on benzene ring and β-diketone moiety. Further antiproliferative activities against five cancer cell lines (Hela, A549, HepG2, MCF-7 and HT-29) in vitro showed that compound 4a-4e displayed remarkable anti-tumor effect against A549, HepG2, MCF-7 and HT-29. Of them, compound 4d is particularly prominent against MCF-7, with IC50 of 1.39 μM. Preliminary mechanism found that compound 4d could trigger ferrous ions and ROS accumulation, increase MDA level in MCF-7 cells, while significantly down-regulate GPX4 level in dose-dependent manner. Western Blot results discovered that compound 4d decreased the ratio of SLC7A11 to GAPDH and GPX4 to β-actin. Docking results indicated that compound 4d had good binding affinity to the active site of GPX4 (PDB ID: 7u4n and 7u4k). In conclusion, compound 4d may be potential anti-tumor agent, which induces ferroptosis in MCF-7 cells through activating SLC7A11/GPX4 axis.
Collapse
Affiliation(s)
- Zhiwen Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, People's Republic of China
| | - Guoqiang Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, People's Republic of China
| | - Yifan Shang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, People's Republic of China
| | - Jiaxin Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, People's Republic of China
| | - Yongqian Liu
- Department of Pharmacy, Guangdong Maternal and Child Health Hospital, Guangzhou 511400, Guangdong, People's Republic of China
| | - Huixian Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, People's Republic of China; Department of Pharmaceutical Equipment, Yangchun Hospital of Traditional Chinese Medicine, Yangchun 529600, Guangdong, People's Republic of China.
| | - Tao Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, People's Republic of China.
| |
Collapse
|
32
|
Zhang Q, Hao X, Sun X, Jia YC, Zhu YY, Yang YX, Zhu BT. 4-Hydroxyestrogen metabolites strongly prevent chemically-induced ferroptotic hepatocyte injury in vitro and in vivo. Eur J Pharmacol 2025; 993:177313. [PMID: 39921062 DOI: 10.1016/j.ejphar.2025.177313] [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: 10/04/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/10/2025]
Abstract
Ferroptosis is a regulated cell death characterized by excessive accumulation of toxic lipid reactive oxygen species (ROS). Ferroptosis is an underlying cause in some human diseases, including the drug-induced liver injury. The present study aims to determine whether 4-hydroxyestrone (4-OH-E1) and 4-hydroxyestradiol (4-OH-E2), two endogenous catechol estrogens, can prevent chemically-induced ferroptotic hepatocyte injury in vitro and in vivo. The induction of ferroptotic cell death by erastin and RSL3 in rat H-4-II-E and human HuH-7 hepatoma cells is used as in vitro models. 4-OH-E1 and 4-OH-E2 each exhibit a strong protection against erastin/RSL3-induced ferroptosis in H-4-II-E hepatoma cells, and they also strongly abrogate erastin/RSL3-induced accumulation of cellular NO, ROS and lipid-ROS. A similar protective effect is observed with 4-OH-E1 and 4-OH-E2 in RSL3-induced ferroptosis in HuH-7 cells. Mechanistically, these two catechol estrogens protect hepatoma cells against chemically-induced ferroptosis mainly through binding to cellular PDI protein with a high affinity, which leads to inhibition of PDI-catalyzed NOS dimerization (activation), thereby preventing the accumulation of cellular NO, ROS and lipid-ROS. In addition, the direct antioxidant activity of these two estrogens may also partially contribute to their cytoprotective effect. In vivo animal studies show that 4-OH-E1 and 4-OH-E2 also have a strong protective effect against acetaminophen-induced liver injury in a mouse model. Together, the results of this study demonstrate that 4-OH-E1 and 4-OH-E2 are endogenous factors with a strong protective activity against chemically-induced hepatocyte injury both in vitro and in vivo.
Collapse
Affiliation(s)
- Qi Zhang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Xiangyu Hao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Xi Sun
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Yi-Chen Jia
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Yan-Yin Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Yong Xiao Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China; Shenzhen Bay Laboratory, Shenzhen, 518172, China.
| |
Collapse
|
33
|
Sun W, Ren H, Chen L, Zhang B, Mei L, Wen J, Zhang Y, Li J, Yan Y, Lai S. TAp73 modulates proliferation and ferroptosis in mammary epithelial cells. Front Cell Dev Biol 2025; 13:1532910. [PMID: 40248352 PMCID: PMC12003338 DOI: 10.3389/fcell.2025.1532910] [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: 11/22/2024] [Accepted: 03/06/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction TAp73, a transcriptionally active isoform of the p73 gene, is essential for epithelial tissue development. Ferroptosis, a regulated form of cell death characterized by lipid peroxidation and reactive oxygen species (ROS) accumulation, has been increasingly studied in recent years. However, its role in epithelial cells and the regulatory function of TAp73 in this context remain poorly understood. Methods We investigated the role of TAp73 in epithelial cell proliferation and ferroptosis using ectopic overexpression and RNA interference approaches. Cell proliferation was assessed through colony formation and DNA synthesis assays. Ferroptosis was induced using RSL3, and the effects were evaluated by measuring cell viability, ROS levels, and the expression of ferroptosis-associated genes PTGS2 and TFRC. Results TAp73 overexpression significantly increased p21 expression, suppressed colony formation and DNA synthesis, thereby inhibiting cell proliferation. In contrast, TAp73 knockdown reduced p21 levels and enhanced cell proliferation. RSL3 treatment induced a dose-dependent increase in cell death and ROS accumulation, confirming the susceptibility of epithelial cells to ferroptosis. Furthermore, TAp73 overexpression enhanced RSL3-induced ferroptosis by upregulating PTGS2 and TFRC, while TAp73 knockdown diminished their expression, reducing oxidative stress and lipid peroxidation. Conclusion TAp73 acts as a dual regulator of epithelial cell fate by inhibiting proliferation and promoting ferroptosis. These findings reveal a novel role for TAp73 in epithelial cell biology and suggest potential therapeutic targets for diseases involving epithelial cell death.
Collapse
Affiliation(s)
- Wenqiang Sun
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| | - Hanjun Ren
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| | - Le Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| | - Bingfei Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| | - Liping Mei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| | - Jiaqi Wen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| | - Yilu Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| | - Jiaqi Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| | - Yongping Yan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| | - Songjia Lai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya’an, China
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Ya’an, China
| |
Collapse
|
34
|
She W, Su J, Ma W, Ma G, Li J, Zhang H, Qiu C, Li X. Natural products protect against spinal cord injury by inhibiting ferroptosis: a literature review. Front Pharmacol 2025; 16:1557133. [PMID: 40248093 PMCID: PMC12003294 DOI: 10.3389/fphar.2025.1557133] [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: 01/08/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Spinal cord injury (SCI) is a severe traumatic condition that frequently results in various neurological disabilities, including significant sensory, motor, and autonomic dysfunctions. Ferroptosis, a recently identified non-apoptotic form of cell death, is characterized by the accumulation of reactive oxygen species (ROS), intracellular iron overload, and lipid peroxidation, ultimately culminating in cell death. Recent studies have demonstrated that ferroptosis plays a critical role in the pathophysiology of SCI, contributing significantly to neural cell demise. Three key cellular enzymatic antioxidants such as glutathione peroxidase 4 (GPX4), ferroptosis suppressor protein 1 (FSP1), and dihydroorotate dehydrogenase (DHODH), have been elucidated as crucial components in the defense against ferroptosis. Natural products, which are bioactive compounds mostly derived from plants, have garnered considerable attention for their potential therapeutic effects. Numerous studies have reported that several natural products can effectively mitigate neural cell death and alleviate SCI symptoms. This review summarizes fifteen natural products containing (-)-Epigallocatechin-3-gallate (EGCG), Proanthocyanidin, Carnosic acid, Astragaloside IV, Trehalose, 8-gingerol, Quercetin, Resveratrol, Albiflorin, Alpha-tocopherol, Celastrol, Hispolon, Dendrobium Nobile Polysaccharide, Silibinin, and Tetramethylpyrazine that have shown promise in treating SCI by inhibiting ferroptosis. Additionally, this review provides an overview of the mechanisms involved in these studies and proposes several perspectives to guide future research directions.
Collapse
Affiliation(s)
- Wei She
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Junxiao Su
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Wenji Ma
- Department of Orthopaedic Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Guohai Ma
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jianfu Li
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Hui Zhang
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Cheng Qiu
- Department of Orthopaedic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xingyong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Department of Orthopaedic Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
35
|
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
|
36
|
Mu F, Luo P, Zhu Y, Nie P, Li B, Bai X. Iron Metabolism and Ferroptosis in Diabetic Kidney Disease. Cell Biochem Funct 2025; 43:e70067. [PMID: 40166850 DOI: 10.1002/cbf.70067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
Diabetic kidney disease (DKD) is a major diabetic microvascular complication that still lacks effective therapeutic drugs. Ferroptosis is a recently identified form of programmed cell death that is triggered by iron overload. It is characterized by unrestricted lipid peroxidation and subsequent membrane damage and is found in various diseases. Accumulating evidence has highlighted the crucial roles of iron overload and ferroptosis in DKD. Here, we review iron metabolism and the biology of ferroptosis. The role of aberrant ferroptosis in inducing diverse renal intrinsic cell death, oxidative stress, and renal fibrosis in DKD is summarized, and we elaborate on critical regulatory factors related to ferroptosis in DKD. Finally, we focused on the significance of ferroptosis in the treatment of DKD and highlight recent data regarding the novel activities of some drugs as ferroptosis inhibitors in DKD, aiming to provide new research targets and treatment strategies on DKD.
Collapse
Affiliation(s)
- Fangxin Mu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Ping Luo
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Yuexin Zhu
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Ping Nie
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Bing Li
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| | - Xue Bai
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Lee WC, Moi SH, Yang SF, Tseng HH, Liu YP. Downregulation of AATK enhances susceptibility to ferroptosis by promoting endosome recycling in gefitinib-resistant lung cancer cells. J Pathol 2025; 265:422-436. [PMID: 39871626 DOI: 10.1002/path.6393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/29/2025]
Abstract
Ferroptosis has been characterised by disruption of the cell membrane through iron-related lipid peroxidation. However, regulation of iron homeostasis in lung cancer cells that are resistant to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) remains unclear. Transcriptome analysis identified a significant downregulation of apoptosis-associated tyrosine kinase (AATK) mRNA expression in gefitinib-resistant PC9 (PC9-GR) cells, which were found to be more susceptible to ferroptosis inducers. An in-depth analysis of publicly available datasets revealed that downregulation of AATK mRNA was associated with lymph node metastasis and poor prognosis in patients with lung adenocarcinoma. Knockdown of AATK-sensitised PC9, HCC827, and H441 cells to the ferroptosis inducer RSL3, whereas ectopic expression of AATK reduced RSL3-induced cell death in PC9-GR and HCC827-GR cells. Compared to PC9 cells, PC9-GR cells exhibited higher transferrin uptake, endosome recycling rate, and increased intracellular iron levels. Blocking iron transport reduced RSL3-induced ferroptosis in PC9-GR cells. Mechanistic studies showed that AATK localised to both early and recycling endosomes. Knockdown of AATK facilitated endosome recycling and elevated intracellular ferrous iron (Fe2+) levels in PC9 cells. Conversely, ectopic expression of AATK delayed endosome recycling and reduced intracellular Fe2+ levels in PC9-GR cells. Inhibition of AATK downregulation-induced iron accumulation decreased RSL3-induced ferroptosis. Taken together, our study indicates that the downregulation of AATK contributes to endosome recycling and iron accumulation, leading to an increased susceptibility to ferroptosis in EGFR-TKI-resistant lung cancer cells. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Wei-Chang Lee
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sin-Hua Moi
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sheau-Fang Yang
- Department of Pathology, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
- Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ho-Hsing Tseng
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Peng Liu
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
38
|
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: 10] [Impact Index Per Article: 10.0] [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
|
39
|
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
|
40
|
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
|
41
|
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
|
42
|
Ma Y, Xu K, Feng J, Zhao X, Tian P, Luo J, Xu L, Song J, Lu C. GSH-Responsive Nano-Photosensitizer for Potentiating Photodynamic Therapy Through Multi-Pronged Synergistic Upregulation of Ferroptosis Sensitivity. Antioxidants (Basel) 2025; 14:407. [PMID: 40298634 PMCID: PMC12024321 DOI: 10.3390/antiox14040407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Impeded by the limited light penetration of photodynamic therapy (PDT) to tissues and the hypoxic environment of solid tumors, the clinical therapeutic efficacy and application are below expectations. In this study, a glutathione (GSH)-responsive nano-photosensitizer, based on the chlorquinaldol (CQD)-loaded iron-containing nanorod composed of meso-tetra (4-carboxyphenyl) porphyrin (TCPP), was prepared to serve as the laser-ignited ferroptosis sensitizer to improve the tumoricidal effect of PDT. In the tumor microenvironment (TME) with elevated GSH levels, therapeutic cargos and ferrous ions are released and are accompanied by the degradation of the nano-photosensitizer and GSH exhaustion. This not only increases liable iron pool (LIP) accumulation by the released ferrous ions but also decreases glutathione peroxidase 4 (GPX4) activity by GSH exhaustion. Simultaneously, GSH exhaustion disrupts intracellular redox homeostasis, heightening NIR light irradiation-triggered photosensitive oxidative stress. Moreover, the released CQD elevates the level of intracellular reactive oxygen species (ROS), enabling the nanorods to gain an oxygen radical generation ability and enhancing the photosensitive oxidative therapeutic efficacy. Strikingly, CQD exacerbates the downregulation of GPX4 expression to promote the accumulation of lipid peroxides. Therefore, we herald a new paradigm for synergistically potentiating PDT based on the "all-in-one" nano-photosensitizer through the multi-pronged upregulation of ferroptosis sensitivity.
Collapse
Affiliation(s)
- Yunong Ma
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (Y.M.); (K.X.); (J.F.); (X.Z.); (P.T.); (J.L.); (L.X.)
| | - Kexin Xu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (Y.M.); (K.X.); (J.F.); (X.Z.); (P.T.); (J.L.); (L.X.)
| | - Jing Feng
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (Y.M.); (K.X.); (J.F.); (X.Z.); (P.T.); (J.L.); (L.X.)
| | - Xi Zhao
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (Y.M.); (K.X.); (J.F.); (X.Z.); (P.T.); (J.L.); (L.X.)
| | - Peilin Tian
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (Y.M.); (K.X.); (J.F.); (X.Z.); (P.T.); (J.L.); (L.X.)
| | - Jiayang Luo
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (Y.M.); (K.X.); (J.F.); (X.Z.); (P.T.); (J.L.); (L.X.)
| | - Luyao Xu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (Y.M.); (K.X.); (J.F.); (X.Z.); (P.T.); (J.L.); (L.X.)
| | - Jiaxing Song
- Cell and Immunology Laboratory, Medical Research Centre, School of Life Sciences and Medical Engineering, Guangxi Medical University, Nanning 530021, China
| | - Cuixia Lu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (Y.M.); (K.X.); (J.F.); (X.Z.); (P.T.); (J.L.); (L.X.)
| |
Collapse
|
43
|
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
|
44
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
45
|
Winkelkotte AM, Al-Shami K, Chaves-Filho AB, Vogel FCE, Schulze A. Interactions of Fatty Acid and Cholesterol Metabolism with Cellular Stress Response Pathways in Cancer. Cold Spring Harb Perspect Med 2025; 15:a041548. [PMID: 38951029 PMCID: PMC11875093 DOI: 10.1101/cshperspect.a041548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Lipids have essential functions as structural components of cellular membranes, as efficient energy storage molecules, and as precursors of signaling mediators. While deregulated glucose and amino acid metabolism in cancer have received substantial attention, the roles of lipids in the metabolic reprogramming of cancer cells are less well understood. However, since the first description of de novo fatty acid biosynthesis in cancer tissues almost 70 years ago, numerous studies have investigated the complex functions of altered lipid metabolism in cancer. Here, we will summarize the mechanisms by which oncogenic signaling pathways regulate fatty acid and cholesterol metabolism to drive rapid proliferation and protect cancer cells from environmental stress. The review also discusses the role of fatty acid metabolism in metabolic plasticity required for the adaptation to changing microenvironments during cancer progression and the connections between fatty acid and cholesterol metabolism and ferroptosis.
Collapse
Affiliation(s)
- Alina M Winkelkotte
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kamal Al-Shami
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Adriano B Chaves-Filho
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Institute of Chemistry, University of São Paulo, 05508000 São Paulo, Brazil
| | - Felix C E Vogel
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
46
|
Hao X, Wang Y, Hou MJ, Liao L, Yang YX, Wang YH, Zhu BT. Raloxifene Prevents Chemically-Induced Ferroptotic Neuronal Death In Vitro and In Vivo. Mol Neurobiol 2025; 62:3934-3955. [PMID: 39354232 PMCID: PMC11790820 DOI: 10.1007/s12035-024-04497-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/12/2024] [Indexed: 10/03/2024]
Abstract
Ferroptosis, a regulated form of cell death characterized by excessive iron-dependent lipid peroxidation, can be readily induced in cultured cells by chemicals such as erastin and RSL3. Protein disulfide isomerase (PDI) has been identified as an upstream mediator of chemically induced ferroptosis and also a target for ferroptosis protection. In this study, we discovered that raloxifene (RAL), a selective estrogen receptor modulator known for its neuroprotective actions in humans, can effectively inhibit PDI function and provide robust protection against chemically induced ferroptosis in cultured HT22 neuronal cells. Specifically, RAL can bind directly to PDI both in vitro and in intact neuronal cells and inhibit its catalytic activity. Computational modeling analysis reveals that RAL can tightly bind to PDI through forming a hydrogen bond with its His256 residue, and biochemical analysis further shows that when PDI's His256 is mutated to Ala256, RAL loses its inhibition of PDI's catalytic activity. This inhibition of PDI by RAL significantly reduces the dimerization of both the inducible and neuronal nitric oxide synthases and the accumulation of nitric oxide, both of which have recently been shown to play a crucial role in mediating chemically induced ferroptosis through subsequent induction of ROS and lipid-ROS accumulation. In vivo behavioral analysis shows that mice treated with RAL are strongly protected against kainic acid-induced memory deficits and hippocampal neuronal damage. In conclusion, this study demonstrates that RAL is a potent inhibitor of PDI and can effectively prevent chemically induced ferroptosis in hippocampal neurons both in vitro and in vivo. These findings offer a novel estrogen receptor-independent mechanism for RAL's neuroprotective actions in animal models and humans.
Collapse
Affiliation(s)
- Xiangyu Hao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Yifan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Ming-Jie Hou
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Lixi Liao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Yong Xiao Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Ying-Hua Wang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China.
- Shenzhen Bay Laboratory, Shenzhen, 518,055, China.
| |
Collapse
|
47
|
Sahoo SS, Manna D. Nanomaterial-Triggered Ferroptosis and Cuproptosis in Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412462. [PMID: 40018870 DOI: 10.1002/smll.202412462] [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: 12/20/2024] [Revised: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Cancer remains one of the leading causes of the death of individuals globally. Conventional treatment techniques like chemotherapy and radiation often suffer various drawbacks like toxicity and drug resistance. The study of cell death has been predominantly focused on classical forms like apoptosis, but the role of metal ions in governing controlled cell death is a fascinating and less explored area. Metal-mediated controlled cell death is a process where metal triggers cell death via a unique mechanism. Nanomaterial-based strategies have gained attention for their ability to deliver precise therapeutic agents while also triggering Regulated Cell Death (RCD) mechanisms in cancer cells. The recently discovered metal-mediated controlled cell death techniques like cuproptosis and ferroptosis can be used in cancer treatment as they can be used selectively for the treatment of drug-resistant cancer. Nano material-based delivery system can also be used for the precise delivery of the drug to the targeted sites. In this review, we have given some idea about the mechanism of metal-mediated controlled cell death techniques (ferroptosis and cuproptosis) and how we can initiate controlled cell deaths using nanomaterials for cancer treatment.
Collapse
Affiliation(s)
- Suman Sekhar Sahoo
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| | - Debasish Manna
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
48
|
Shen Y, Zhang G, Wei C, Zhao P, Wang Y, Li M, Sun L. Potential role and therapeutic implications of glutathione peroxidase 4 in the treatment of Alzheimer's disease. Neural Regen Res 2025; 20:613-631. [PMID: 38886929 PMCID: PMC11433915 DOI: 10.4103/nrr.nrr-d-23-01343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/27/2023] [Accepted: 12/21/2023] [Indexed: 06/20/2024] Open
Abstract
Alzheimer's disease is an age-related neurodegenerative disorder with a complex and incompletely understood pathogenesis. Despite extensive research, a cure for Alzheimer's disease has not yet been found. Oxidative stress mediates excessive oxidative responses, and its involvement in Alzheimer's disease pathogenesis as a primary or secondary pathological event is widely accepted. As a member of the selenium-containing antioxidant enzyme family, glutathione peroxidase 4 reduces esterified phospholipid hydroperoxides to maintain cellular redox homeostasis. With the discovery of ferroptosis, the central role of glutathione peroxidase 4 in anti-lipid peroxidation in several diseases, including Alzheimer's disease, has received widespread attention. Increasing evidence suggests that glutathione peroxidase 4 expression is inhibited in the Alzheimer's disease brain, resulting in oxidative stress, inflammation, ferroptosis, and apoptosis, which are closely associated with pathological damage in Alzheimer's disease. Several therapeutic approaches, such as small molecule drugs, natural plant products, and non-pharmacological treatments, ameliorate pathological damage and cognitive function in Alzheimer's disease by promoting glutathione peroxidase 4 expression and enhancing glutathione peroxidase 4 activity. Therefore, glutathione peroxidase 4 upregulation may be a promising strategy for the treatment of Alzheimer's disease. This review provides an overview of the gene structure, biological functions, and regulatory mechanisms of glutathione peroxidase 4, a discussion on the important role of glutathione peroxidase 4 in pathological events closely related to Alzheimer's disease, and a summary of the advances in small-molecule drugs, natural plant products, and non-pharmacological therapies targeting glutathione peroxidase 4 for the treatment of Alzheimer's disease. Most prior studies on this subject used animal models, and relevant clinical studies are lacking. Future clinical trials are required to validate the therapeutic effects of strategies targeting glutathione peroxidase 4 in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Yanxin Shen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Guimei Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Chunxiao Wei
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Panpan Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Yongchun Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Mingxi Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
- Cognitive Impairment Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
49
|
Chen SY, Shyu IL, Chi JT. NINJ1 in Cell Death and Ferroptosis: Implications for Tumor Invasion and Metastasis. Cancers (Basel) 2025; 17:800. [PMID: 40075648 PMCID: PMC11898531 DOI: 10.3390/cancers17050800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
NINJ1 was initially recognized for its role in nerve regeneration and cellular adhesion. Subsequent studies have uncovered its participation in cancer progression, where NINJ1 regulates critical steps in tumor metastasis, such as cell migration and invasion. More recently, NINJ1 has emerged as a multifunctional protein mediating plasma membrane rupture (PMR) in several lytic cell death processes, including apoptosis, necroptosis, and pyroptosis. However, its role in ferroptosis-an iron-dependent form of lytic cell death characterized by lipid peroxidation-remained unclear until 2024. Ferroptosis is a tumor suppression mechanism that may be particularly relevant to detached and metastatic cancer cells. This review explores the role of NINJ1 in tumor invasion and metastasis, focusing on its regulation of ferroptosis via a non-canonical mechanism distinct from other cell deaths. We discuss the process of ferroptosis and its implications for cancer invasion and metastasis. Furthermore, we review recent studies highlighting the diverse roles of NINJ1 in ferroptosis regulation, including its canonical function in PMR and its non-canonical function of modulating intracellular levels of glutathione (GSH) and coenzyme A (CoA) via interaction with xCT anti-porter. Given that ferroptosis has been associated with tumor suppression, metastasis, the elimination of treatment-resistant cancer cells, and tumor dormancy, NINJ1's modulation of ferroptosis presents a promising therapeutic target for inhibiting metastasis. Understanding the dual role of NINJ1 in promoting or restraining ferroptosis depending on cellular context could open avenues for novel anti-cancer strategies to enhance ferroptotic vulnerability in metastatic tumors.
Collapse
Affiliation(s)
- Ssu-Yu Chen
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA;
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ing-Luen Shyu
- Department of Obstetrics and Gynecology, Chi Mei Medical Center, Tainan 710, Taiwan
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Center for Advanced Genomic Technologies, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
50
|
Ding Z, Li Z, Sun K, Liu Y, Fang Z, Sun S, Li C, Wang Z. Mitochondrial Regulation of Ferroptosis in Cancer Cells. Int J Biol Sci 2025; 21:2179-2200. [PMID: 40083691 PMCID: PMC11900798 DOI: 10.7150/ijbs.105446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Ferroptosis is an iron-dependent nonapoptotic regulated cell death modality characterized by lethal levels of lipid peroxide accumulation and disrupted antioxidant systems. An increasing number of studies have revealed correlations between ferroptosis and the pathophysiology and treatment of cancer. Given the intricate involvement of mitochondria in ferroptosis, as suggested by previous studies, here, we review advances in understanding the roles of mitochondrial quality control and mitochondrial metabolism (including the roles of the TCA cycle, reactive oxygen species, iron metabolism, and lipid metabolism) in cancer-related ferroptosis and outline the molecular mechanism and clinical translation of mitochondria-related ferroptosis in cancer treatment. with the aim of promoting the precise utilization and prevention of ferroptosis in cancer therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | - Shengrong Sun
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Chenyuan Li
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Zhong Wang
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| |
Collapse
|