1
|
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
|
2
|
Kandettu A, Ghosal J, Tharayil JS, Kuthethur R, Mallya S, Narasimhamurthy RK, Mumbrekar KD, Subbannayya Y, Kumar NA, Radhakrishnan R, Kabekkodu SP, Chakrabarty S. Inhibition of mitochondrial genome-encoded mitomiR-3 contributes to ZEB1 mediated GPX4 downregulation and pro-ferroptotic lipid metabolism to induce ferroptosis in breast cancer cells. Free Radic Biol Med 2025; 234:151-168. [PMID: 40239722 DOI: 10.1016/j.freeradbiomed.2025.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/04/2025] [Accepted: 04/12/2025] [Indexed: 04/18/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, represents a unique vulnerability in cancer cells. However, current ferroptosis-inducing therapies face clinical limitations due to poor cancer cell specificity, systemic toxicity, and off-target effects. Therefore, a deeper understanding of molecular regulators of ferroptosis sensitivity is critical for developing targeted therapies. The metabolic plasticity of cancer cells determines their sensitivity to ferroptosis. While mitochondrial dysfunction contributes to metabolic reprogramming in cancer, its role in modulating ferroptosis remains poorly characterized. Previously, studies have identified that mitochondrial genome also encodes several non-coding RNAs. We identified 13 novel mitochondrial genome-encoded miRNAs (mitomiRs) that are aberrantly overexpressed in triple-negative breast cancer (TNBC) cell lines and patient tumors. We observed higher levels of mitomiRs in basal-like triple-negative breast cancer (TNBC) cells compared to mesenchymal stem-like TNBC cells. Strikingly, 11 of these mitomiRs directly target the 3'UTR of ZEB1, a master regulator of epithelial-to-mesenchymal transition (EMT). Using mitomiR-3 mimic, inhibitor and sponges, we demonstrated its role as a key regulator of ZEB1 expression in TNBC cells. Inhibition of mitomiR-3 via sponge construct in basal-like TNBC, MDA-MB-468 cells, promoted ZEB1 upregulation and induced a mesenchymal phenotype. Further, mitomiR-3 inhibition in TNBC cells contributed to reduced cancer cell proliferation, migration, and invasion. Mechanistically, mitomiR-3 inhibition in TNBC cells promote metabolic reprogramming toward pro-ferroptotic pathways, including iron accumulation, increased polyunsaturated fatty acid (PUFA) metabolites, and lipid peroxidation, contributing to ferroptotic cell death via ZEB1-mediated downregulation of GPX4, a critical ferroptosis defense enzyme. We observed that mitomiR-3 inhibition significantly suppressed tumor growth in vivo. Our identified mitomiR-3 has low expression in normal breast cells, minimizing potential off-target toxicity, making them a promising target for pro-ferroptotic cancer therapy. Our study reveals a novel link between mitochondrial miRNAs and ferroptosis sensitivity in TNBC paving a way for miRNA-based therapeutics.
Collapse
Affiliation(s)
- Amoolya Kandettu
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Joydeep Ghosal
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Jesline Shaji Tharayil
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Raviprasad Kuthethur
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sandeep Mallya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rekha Koravadi Narasimhamurthy
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Kamalesh Dattaram Mumbrekar
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Yashwanth Subbannayya
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Naveena An Kumar
- Department of Surgical Oncology, Manipal Comprehensive Cancer Care Centre, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India; Academic Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, S10TTA, UK; Academic Unit of Oral Biology and Oral Pathology, Oman Dental College, Wattayah 116, Muscat, Oman
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Sanjiban Chakrabarty
- Department of Public Health Genomics, Centre for DNA Repair and Genome Stability (CDRGS), Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
3
|
Shen Y, Wei W, Lu Y, Song M, Yang S, Liu H, Xu X, Zhou H. Berberine alleviates tendinopathy by suppressing the cGAS-STING pathway and Relieving ferroptosis. Biochem Biophys Res Commun 2025; 767:151923. [PMID: 40319817 DOI: 10.1016/j.bbrc.2025.151923] [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] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Berberine, a key bioactive component of Coptis rhizome, has been extensively studied for its therapeutic effects on various diseases. This research aimed to investigate the potential benefits of berberine in treating tendinopathy and to elucidate the underlying mechanisms through animal and laboratory studies. Our findings indicated that berberine effectively treated type I collagenase-induced tendinopathy in rats, confirmed by cellular-level validation. At the molecular level, berberine reduced the activation of the cGAS-STING signaling pathway and decreased the accumulation of malondialdehyde (MDA) and reactive oxygen species (ROS) in both animal models and cell cultures. Additionally, berberine upregulated the expression of glutathione (GSH) and glutathione peroxidase 4 (GPX4) in tissues. These results suggested that berberine alleviated ferroptosis via the cGAS-STING pathway, thus exerting therapeutic effects on tendinopathy. To validate these findings further, we administered the ferroptosis inducer Imidazole Ketone Erastin (IKE) to evaluate the effects of berberine. IKE significantly diminished the therapeutic effects of berberine on tendinopathy, as indicated by the previously mentioned markers. Thus, berberine mitigated ferroptosis by inhibiting the cGAS-STING pathway, highlighting its potential in managing tendinopathy.
Collapse
Affiliation(s)
- Yeshuai Shen
- The Second Affiliated Hospital of Soochow University, China; The Affiliated Hospital of Xuzhou Medical University, China
| | - Wenting Wei
- The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, China
| | - Yang Lu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | | | - Shaojie Yang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Huan Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xingquan Xu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| | - Haibin Zhou
- The Second Affiliated Hospital of Soochow University, China.
| |
Collapse
|
4
|
Gao D, Chen T, Dong J, He Y, Ge N, Guo J, Sun S, Yang F. Ferroptosis at the crossroads: Insights and advances in non-neoplastic pancreatic diseases. Int Immunopharmacol 2025; 158:114870. [PMID: 40383100 DOI: 10.1016/j.intimp.2025.114870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/26/2025] [Accepted: 05/10/2025] [Indexed: 05/20/2025]
Abstract
Ferroptosis is a form of regulated cell death characterized by iron accumulation and increased lipid peroxidation, primarily counteracted by a range of antioxidant molecules, including glutathione (GSH), glutathione peroxidase 4 (GPX4), ubiquinone, tetrahydrofolate, and nuclear respiratory factor 2. Furthermore, the process of ferroptosis is intricately influenced by the opposing actions of the p53 tumor suppressor gene and activated transcription factors 3 and 4, which can either facilitate or hinder ferroptotic cell death depending on the cellular context. This form of cell death is significantly associated with various pancreatic disorders, including both acute and chronic pancreatitis, as well as diabetes mellitus. In this review, we thoroughly investigate the mechanisms underlying ferroptosis, focusing on iron overload, lipid peroxidation, and the regulatory molecules involved in ferroptosis modulation (notably the system xc-/GSH/GPX4 axis), along with the relevant signaling pathways. We also examine the role of ferroptosis in non-neoplastic pancreatic diseases such as pancreatitis and diabetes mellitus while identifying novel therapeutic agents that target ferroptosis, potentially paving the way for innovative treatment strategies for pancreatic conditions.
Collapse
Affiliation(s)
- Duolun Gao
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Tingting Chen
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Jize Dong
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Yanjie He
- Department of Surgery, New York University School of Medicine and NYU-Langone Medical Center, New York 10012, NY, USA
| | - Nan Ge
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Jintao Guo
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China
| | - Siyu Sun
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China.
| | - Fan Yang
- Department of Gastroenterology, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, People's Republic of China.
| |
Collapse
|
5
|
Li MH, Yang Y, Dong QQ, Tao H, Lu C, Yang JJ. Novel epitranscriptomic and epigenetic therapeutic strategies and targets for ferroptosis in liver fibrosis. Eur J Pharmacol 2025; 996:177344. [PMID: 40015597 DOI: 10.1016/j.ejphar.2025.177344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
Liver fibrosis is characterized by an excessive accumulation of extracellular matrix (ECM) and the activation of hepatic stellate cells (HSCs), which are influenced by epitranscriptomic and epigenetic factors. Recent advancements in epigenetic and epitranscriptomic research have revealed new opportunities for therapeutic interventions, particularly through the regulation of ferroptosis, a type of programmed cell death that is specifically linked to iron-dependent lipid peroxidation. In the context of liver fibrosis, a progressive scarring process that can progress to cirrhosis and ultimately end-stage liver disease, targeting these regulatory mechanisms to modulate ferroptosis presents a promising therapeutic strategy. This review aims to consolidate current knowledge on the epigenetic and epitranscriptomic control of ferroptosis and investigate its potential implications for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ming-Hui Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yang Yang
- Department of General Surgery, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
| | - Qi-Qi Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Chao Lu
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
6
|
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
|
7
|
Sun X, Teng R, Xu N, Sun Y, Zhang E, Chen X, Guo Q, Li S. PFOS exposure impairs porcine oocyte maturation and embryo development via mitochondria-dependent ferroptosis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 374:126185. [PMID: 40189092 DOI: 10.1016/j.envpol.2025.126185] [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: 01/03/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
Perfluorooctane sulfonate (PFOS) is a widely utilized chemical known for its exceptional environmental stability over extended periods, its significant potential to bioaccumulate in living organisms, and its considerable risks to both health and the environment. Several studies have suggested that PFOS may pose reproductive risks in mammals; however, the exact mechanisms driving these effects are not well understood. In this study, we explored the possible mechanisms by which PFOS toxicity affects the maturation of mammalian oocytes and the embryonic development employing porcine oocytes as a model system. SMART-seq results suggested that PFOS may affect oocyte maturation through mechanisms involving ferroptosis, autophagy, and alterations in membrane structure. Our results suggest that PFOS exposure adversely affects mitochondrial function and structure, thereby influencing peroxisome biogenesis and contributing to oxidative stress. Most importantly, we found that exposure to PFOS significantly elevated Fe2+ levels, an indicator associated with ferroptosis in oocytes. Furthermore, malondialdehyde (MDA) levels in the PFOS group were significantly higher than those in the control group. Additionally, the mRNA expression levels of PCBP1 and PCBP2, which are related to ferroptosis, as well as the expression level of P53, were significantly reduced in the PFOS group. Overall, exposure to PFOS in vitro results in mitochondrial damage in porcine oocytes, which induces lipid peroxidation and subsequently leads to the occurrence of ferroptosis.
Collapse
Affiliation(s)
- Xiaoqing Sun
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Ran Teng
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Ning Xu
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Yutong Sun
- Affiliated Middle School to Jilin University, Changchun, 130000, China
| | - Enbo Zhang
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Xingfu Chen
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China
| | - Qing Guo
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Suo Li
- Jilin Agricultural University, Xincheng Street 2888, Changchun, 130118, China.
| |
Collapse
|
8
|
Saadh MJ, Ahmed HH, Sanghvi G, Bin Awang Isa MZ, Singh P, Kaur K, Kumar MR, Husseen B. Recent advances in the delivery of microRNAs via exosomes derived from MSCs, and their role in regulation of ferroptosis. Pathol Res Pract 2025; 270:155984. [PMID: 40315562 DOI: 10.1016/j.prp.2025.155984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025]
Abstract
Mesenchymal stem cell (MSC) therapy, with its unique properties, has garnered interest in cancer treatment. Exosomes (EXOs)-derived from MSC retain the paracrine components of MSCs and demonstrate increased stability, minimal immunogenicity, and low risk of unintended tumorigenesis. Enhanced endocytosis methods make them versatile delivery vehicles for therapeutic cargo. MSC-EXOs can either promote or inhibit carcinogenesis, mediated by paracrine factors and various RNA molecules, particularly microRNAs (miRNAs). The prospect of using MSC-EXOs as a delivery tool for antitumor miRNAs in solid tumor therapy is promising. Exosomes' intrinsic tumor-targeting abilities and low immunogenicity make them ideal for delivering miRNAs, which have shown potential as cancer therapeutics. miRNAs within MSC-EXOs molecules can stimulate tumor growth or induce non-apoptotic cell death pathways, such as ferroptosis, depending on context. Ferroptosis is a kind of controlled cell death that is associated with the pathophysiology of several illnesses and includes iron metabolism. There is growing evidence that miRNAs carried by exosomes derived from MSCs may control ferroptosis in tumor cells by altering key genes related to antioxidant defense, lipid peroxidation, and iron metabolism. Understanding their complex mechanisms in the tumor microenvironment and optimizing their cargo are critical steps toward harnessing their full therapeutic potential. This review provides a comprehensive overview of MSC-EXOs and their role in cancer treatment. We also discuss the potential of MSC-EXOs as delivery vehicles for miRNAs to enhance therapeutic efficacy, as well as the role of exosomal miRNAs in the induction of ferroptosis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | | | - Priyanka Singh
- NIMS School of Allied Sciences and Technology, NIMS University, Jaipur, Rajasthan 303121, India
| | - Kiranjeet Kaur
- Chandigarh Pharmacy College, Chandigarh Group of colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Beneen Husseen
- Medical laboratory technique college, the Islamic University, Najaf, Iraq; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
9
|
Meng J, Wen C, Lu Y, Fan X, Dang R, Chu J, Jiang P, Han W, Feng L. Alliin from garlic as a neuroprotective agent attenuates ferroptosis in vitro and in vivo via inhibiting ALOX15. Food Funct 2025. [PMID: 40396992 DOI: 10.1039/d5fo00425j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Alliin, a precursor active compound of sulfur-containing organic compounds such as allicin in garlic, is recognized as an important bioactive substance in garlic. Allicin has been shown to have significant neuroprotective effects and promote functional recovery in intracerebral hemorrhage (ICH). As a precursor of many active compounds, alliin may have broader therapeutic effects. Therefore, the aim of this study was to investigate the molecular mechanisms underlying the neuroprotective effects of alliin. In this study, we found that alliin inhibits ferroptosis, thereby exerting neuroprotective effects in ICH. However, the neuroprotective effects of alliin and its pharmacological mechanisms in ferroptosis have not been fully explored. The results showed that alliin significantly inhibited erastin-induced ferroptosis in HT22 cells and suppressed ferroptosis in the brain tissue of collagenase-induced ICH mice, alleviating neurological dysfunction and pathological damage. Mechanistically, alliin downregulated the expression of 15-lipoxygenase (ALOX15), which inhibits phospholipid peroxidation and ferroptosis. Moreover, gene knockout of ALOX15 produced effects similar to those of alliin, and comparable results were obtained using the ferroptosis inhibitor ferrostatin-1. This study is the first to demonstrate that alliin regulates ferroptosis both in vitro and in vivo. In conclusion, our study highlights ALOX15 as a critical factor in ferroptosis associated with ICH, and shows that alliin exerts neuroprotective effects by inhibiting ALOX15-dependent ferroptosis.
Collapse
Affiliation(s)
- Junjun Meng
- Translational Pharmaceutical Laboratory, Jining NO.1 People's Hospital, Shandong First Medical University, Jiankang Road, Jining 272000, China
- Shandong Provincial Key Medical and Health Laboratory of Neuroinjury and Repair, Jining NO.1 People's Hospital, Jining, 272000, China
| | - Chengquan Wen
- Department of Pharmacy, Qingdao Eighth People's Hospital, China
| | - Yang Lu
- Clinical College of Jining Medical University, China
| | - Xiaofan Fan
- Shandong Provincial Key Medical and Health Laboratory of Neuroinjury and Repair, Jining NO.1 People's Hospital, Jining, 272000, China
| | - Ruili Dang
- Translational Pharmaceutical Laboratory, Jining NO.1 People's Hospital, Shandong First Medical University, Jiankang Road, Jining 272000, China
- Shandong Provincial Key Medical and Health Laboratory of Neuroinjury and Repair, Jining NO.1 People's Hospital, Jining, 272000, China
| | - Jianfeng Chu
- Department of Neurology, Jining No. 1 People's Hospital, Jining, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining NO.1 People's Hospital, Shandong First Medical University, Jiankang Road, Jining 272000, China
- Shandong Provincial Key Medical and Health Laboratory of Neuroinjury and Repair, Jining NO.1 People's Hospital, Jining, 272000, China
| | - Wenxiu Han
- Translational Pharmaceutical Laboratory, Jining NO.1 People's Hospital, Shandong First Medical University, Jiankang Road, Jining 272000, China
- Shandong Provincial Key Medical and Health Laboratory of Neuroinjury and Repair, Jining NO.1 People's Hospital, Jining, 272000, China
| | - Lei Feng
- Department of Neurosurgery, Jining NO.1 People's Hospital, Jining, China.
| |
Collapse
|
10
|
Liu X, Luo Q, Zhao Y, Ren P, Jin Y, Zhou J. The Ferroptosis-Mitochondrial Axis in Depression: Unraveling the Feedforward Loop of Oxidative Stress, Metabolic Homeostasis Dysregulation, and Neuroinflammation. Antioxidants (Basel) 2025; 14:613. [PMID: 40427494 PMCID: PMC12108521 DOI: 10.3390/antiox14050613] [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: 04/11/2025] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Emerging evidence links ferroptosis-mitochondrial dysregulation to depression pathogenesis through an oxidative stress-energy deficit-neuroinflammation cycle driven by iron overload. This study demonstrates that iron accumulation initiates ferroptosis via Fenton reaction-mediated lipid peroxidation, compromising neuronal membrane integrity and disabling the GPx4 antioxidant system. Concurrent mitochondrial complex I/IV dysfunction impairs ATP synthesis, creating an AMPK/mTOR signaling imbalance and calcium dyshomeostasis that synergistically impair synaptic plasticity. Bidirectional crosstalk emerges: lipid peroxidation derivatives oxidize mitochondrial cardiolipin, while mitochondrial ROS overproduction activates ACSL4 to amplify ferroptotic susceptibility, forming a self-reinforcing neurodegenerative loop. Prefrontal-hippocampal metabolomics reveal paradoxical metabolic reprogramming with glycolytic compensation suppressing mitochondrial biogenesis (via PGC-1α/TFAM downregulation), trapping neurons in bioenergetic crisis. Clinical data further show that microglial M1 polarization through cGAS-STING activation sustains neuroinflammation via IL-6/TNF-α release. We propose a "ferroptosis-mitochondrial fragmentation-metabolic maladaptation" triad as mechanistic subtyping criteria for depression. Preclinical validation shows that combinatorial therapy (iron chelators + SIRT3 agonists) rescues neuronal viability by restoring mitochondrial integrity and energy flux. This work shifts therapeutic paradigms from monoaminergic targets toward multimodal strategies addressing iron homeostasis, organelle dynamics, and metabolic vulnerability-a framework with significant implications for developing neuroprotective antidepressants.
Collapse
Affiliation(s)
- Xu Liu
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Qiang Luo
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Yulong Zhao
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Peng Ren
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Yu Jin
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
| | - Junjie Zhou
- School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China; (X.L.); (Q.L.); (Y.Z.); (P.R.); (Y.J.)
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 341000, China
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou 341000, China
| |
Collapse
|
11
|
Karmaker S, Rosales PD, Tirumuruhan B, Viravalli A, Boehnke N. More than a delivery system: the evolving role of lipid-based nanoparticles. NANOSCALE 2025; 17:11864-11893. [PMID: 40293317 DOI: 10.1039/d4nr04508d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Lipid-based nanoparticles, including liposomes and lipid nanoparticles (LNPs), make up an important class of drug delivery systems. Their modularity enables encapsulation of a wide range of therapeutic cargoes, their ease of functionalization allows for incorporation of targeting motifs and anti-fouling coatings, and their scalability facilitates rapid translation to the clinic. While the discovery and early understanding of lipid-based nanoparticles is heavily rooted in biology, formulation development has largely focused on materials properties, such as how liposome and lipid nanoparticle composition can be altered to maximize drug loading, stability and circulation. To achieve targeted delivery and enable improved accumulation of therapeutics at target tissues or disease sites, emphasis is typically placed on the use of external modifications, such as peptide, protein, and polymer motifs. However, these approaches can increase the complexity of the nanocarrier and complicate scale up. In this review, we focus on how our understanding of lipid structure and function in biological contexts can be used to design intrinsically functional and targeted nanocarriers. We highlight formulation-based strategies, such as the incorporation of bioactive lipids, that have been used to modulate liposome and lipid nanoparticle properties and improve their functionality while retaining simple nanocarrier designs. We also highlight classes of naturally occurring lipids, their functions, and how they have been incorporated into lipid-based nanoparticles. We will additionally position these approaches into the historical context of both liposome and LNP development.
Collapse
Affiliation(s)
- Senjuti Karmaker
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Plinio D Rosales
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Barath Tirumuruhan
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Amartya Viravalli
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Natalie Boehnke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| |
Collapse
|
12
|
Zhuang X, Shi S, Liu S, Jiao Y, Huang B, Yang Y, Yang L, Yang X, Wang H, Liang C, Song D, Yu H, Zou D, Sun Q, Yang S, Yin C, Li J, Liu Y, Min J, Wang F, Nian Y, Du L, Chu B. Dipyridamole Acts as Clinical Ferroptosis Inhibitor to Prevent from Tissue Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500566. [PMID: 40365742 DOI: 10.1002/advs.202500566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/18/2025] [Indexed: 05/15/2025]
Abstract
Ferroptosis is a newly identified cell death triggered by iron-induced lipid peroxidation. Numerous studies reveal that ferroptosis participates in multiple types of tissue injury including ischaemia-reperfusion (I/R) injury and doxorubicin (Dox)-induced damage. Targeting ferroptosis is a promising approach for disease treatment as the blockade of ferroptosis efficiently alleviates the symptoms. However, no known ferroptosis inhibitors have been used for clinical treatment. Although certain clinical compounds act as ferroptosis inhibitors in vitro, whether these drugs cure tissue injury by suppressing ferroptosis is little known. Here, by screening a large panel of drugs used in the clinic, it is identified that dipyridamole significantly attenuates Dox or I/R-induced cardiac injury. Moreover, dipyridamole can achieve a good therapeutic effect on liver and kidney injury. Mechanistically, dipyridamole-mediated ferroptosis inhibition is strictly dependent on solute carrier family 7 member 11 (SLC7A11). Dipyridamole down-regulates the expression of ring finger protein 126 (RNF126), which is an E3 ligase to ubiquitinate SLC7A11 for proteasome degradation. Deficiency of SLC7A11 largely blocks the protective role of dipyridamole in vitro and in vivo. Together, the findings uncover that dipyridamole acts as a clinical compound to alleviate organ injury via suppressing ferroptosis, providing novel insights into the clinical therapy for ferroptosis-related tissue damage.
Collapse
Affiliation(s)
- Xiao Zhuang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuang Shi
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, 250012, China
| | - Shuo Liu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yaqiong Jiao
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Bin Huang
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China
| | - Yinghong Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Li Yang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Xinquan Yang
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hui Wang
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China
| | - Chunhui Liang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dandan Song
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huaxiang Yu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Dan Zou
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Qi Sun
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, 250012, China
| | - Shu Yang
- College of Pharmacy, Nanjing drum tower hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chengqian Yin
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China
| | - Jian Li
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, 250012, China
| | - Yiming Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fudi Wang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yong Nian
- College of Pharmacy, Nanjing drum tower hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lutao Du
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China
- Shandong Provincial Key Laboratory of Innovation Technology in Laboratory Medicine, Jinan, 250012, China
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| |
Collapse
|
13
|
Zhang Z, Ma X, Pan W, Li X, Li Y, Fang N, Yin X, Yang L, Hou Q, Yang D, Ji T, Zheng R, Lin M. A Novel Compound 3a-M1, from Metabolites of Sinomenine Derivative 3a, Exerts Potent Anti-Aplastic Anemia Activity via IP 3R/ORAI-Mediated CTL Ferroptosis. J Med Chem 2025; 68:9723-9740. [PMID: 40243551 DOI: 10.1021/acs.jmedchem.5c00546] [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: 04/18/2025]
Abstract
Aplastic anemia (AA) is a refractory hematological disease with limited therapeutic effectiveness and serious treatment-related side effects. Cytotoxic T lymphocytes (CTLs) play a key role in AA pathogenesis. In our previous study, sinomenine derivative 3a was obtained, which demonstrated potential anti-AA activity by targeting CTLs with low toxicity. In this study, a novel metabolite, 3a-M1, was identified with optimized bioavailability from 3a metabolism, which exhibited a more notable effect in alleviating anemia symptoms, suppressing bone marrow CTLs activation, and improving hematopoietic function in immune-mediated bone marrow failure mouse models. In vitro experiments demonstrated that 3a-M1 directly inhibited CTLs activation and their killing function; the underlying mechanism was at least in part mediated by the selective ferroptosis of overactivated CTLs via the IP3R/ORAI pathway. These findings suggest that 3a-M1 represents a novel potential therapeutic agent for AA treatment and ferroptosis may serve as a promising target on CTLs for AA therapy.
Collapse
Affiliation(s)
- Ziqian Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xilan Ma
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Wei Pan
- Center of Information, National Medical Products Administration, Beijing 100044, People's Republic of China
| | - Xiaoshun Li
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yanyan Li
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Nan Fang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xin Yin
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Longlong Yang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Qi Hou
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Dezhi Yang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Tengfei Ji
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Ruifang Zheng
- Xinjiang Key Laboratory of Uygur Medical Research, Xinjiang Institute of Materia Medica, Urumqi 841100, People's Republic of China
| | - Mingbao Lin
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| |
Collapse
|
14
|
Cañeque T, Baron L, Müller S, Carmona A, Colombeau L, Versini A, Solier S, Gaillet C, Sindikubwabo F, Sampaio JL, Sabatier M, Mishima E, Picard-Bernes A, Syx L, Servant N, Lombard B, Loew D, Zheng J, Proneth B, Thoidingjam LK, Grimaud L, Fraser CS, Szylo KJ, Der Kazarian E, Bonnet C, Charafe-Jauffret E, Ginestier C, Santofimia-Castaño P, Estaras M, Dusetti N, Iovanna JL, Cunha AS, Pittau G, Hammel P, Tzanis D, Bonvalot S, Watson S, Gandon V, Upadhyay A, Pratt DA, Freitas FP, Friedmann Angeli JP, Stockwell BR, Conrad M, Ubellacker JM, Rodriguez R. Activation of lysosomal iron triggers ferroptosis in cancer. Nature 2025:10.1038/s41586-025-08974-4. [PMID: 40335696 DOI: 10.1038/s41586-025-08974-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 04/03/2025] [Indexed: 05/09/2025]
Abstract
Iron catalyses the oxidation of lipids in biological membranes and promotes a form of cell death called ferroptosis1. Defining where this chemistry occurs in the cell can inform the design of drugs capable of inducing or inhibiting ferroptosis in various disease-relevant settings. Genetic approaches have revealed suppressors of ferroptosis2-4; by contrast, small molecules can provide spatiotemporal control of the chemistry at work5. Here we show that the ferroptosis inhibitor liproxstatin-1 exerts cytoprotective effects by inactivating iron in lysosomes. We also show that the ferroptosis inducer RSL3 initiates membrane lipid oxidation in lysosomes. We designed a small-molecule activator of lysosomal iron-fentomycin-1-to induce the oxidative degradation of phospholipids and ultimately ferroptosis. Fentomycin-1 is able to kill iron-rich CD44high primary sarcoma and pancreatic ductal adenocarcinoma cells, which can promote metastasis and fuel drug tolerance. In such cells, iron regulates cell adaptation6,7 while conferring vulnerability to ferroptosis8,9. Sarcoma cells exposed to sublethal doses of fentomycin-1 acquire a ferroptosis-resistant cell state characterized by the downregulation of mesenchymal markers and the activation of a membrane-damage response. This phospholipid degrader can eradicate drug-tolerant persister cancer cells in vitro and reduces intranodal tumour growth in a mouse model of breast cancer metastasis. Together, these results show that control of iron reactivity confers therapeutic benefits, establish lysosomal iron as a druggable target and highlight the value of targeting cell states10.
Collapse
Affiliation(s)
- Tatiana Cañeque
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Leeroy Baron
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Sebastian Müller
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Alanis Carmona
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Ludovic Colombeau
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Antoine Versini
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Stéphanie Solier
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
- Department of Genetics, Institut Curie, Paris, France
- Paris Saclay University, UVSQ, Montigny-le-Bretonneux, France
| | - Christine Gaillet
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | | | - Julio L Sampaio
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France
| | - Marie Sabatier
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Laurène Syx
- Institut Curie, INSERM, Mines Paris Tech, PSL Research University, Paris, France
| | - Nicolas Servant
- Institut Curie, INSERM, Mines Paris Tech, PSL Research University, Paris, France
| | | | - Damarys Loew
- Institut Curie, PSL Research University, Paris, France
| | - Jiashuo Zheng
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Laurence Grimaud
- Ecole Normale Supérieure, CNRS, PSL Research University, Paris, France
| | - Cameron S Fraser
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Krystina J Szylo
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Emma Der Kazarian
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Caroline Bonnet
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | | | - Christophe Ginestier
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Patricia Santofimia-Castaño
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Matias Estaras
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Nelson Dusetti
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Juan Lucio Iovanna
- CRCM, CNRS, INSERM, Institut Paoli-Calmettes, Aix-Marseille University, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Antonio Sa Cunha
- Department of Digestive and Medical Oncology, Paris Saclay University and Paul-Brousse Hospital (APHP Sud), Villejuif, France
| | - Gabriella Pittau
- Department of Digestive and Medical Oncology, Paris Saclay University and Paul-Brousse Hospital (APHP Sud), Villejuif, France
| | - Pascal Hammel
- Department of Digestive and Medical Oncology, Paris Saclay University and Paul-Brousse Hospital (APHP Sud), Villejuif, France
| | - Dimitri Tzanis
- Department of Surgical Oncology, Institut Curie, Paris, France
| | - Sylvie Bonvalot
- Department of Surgical Oncology, Institut Curie, Paris, France
| | - Sarah Watson
- Department of Medical Oncology, Institut Curie, INSERM, PSL Research University, Paris, France
| | - Vincent Gandon
- Institut de Chimie Moléculaire et des Matériaux d'Orsay, CNRS, Paris Saclay University, Orsay, France
| | - Aditya Upadhyay
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Florêncio Porto Freitas
- Rudolf-Virchow-Zentrum, Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - José Pedro Friedmann Angeli
- Rudolf-Virchow-Zentrum, Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Brent R Stockwell
- Department of Chemistry, Columbia University, New York, NY, USA
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, TUM Natural School of Sciences, Technical University of Munich, Garching, Germany
| | - Jessalyn M Ubellacker
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Raphaël Rodriguez
- Institut Curie, CNRS, INSERM, PSL Research University, Paris, France.
| |
Collapse
|
15
|
Wei X, Fan X, Chai W, Xiao J, Zhao J, He A, Tang X, Li F, Guo S. Dietary limonin ameliorates heart failure with preserved ejection fraction by targeting ferroptosis via modulation of the Nrf2/SLC7A11/GPX4 axis: an integrated transcriptomics and metabolomics analysis. Food Funct 2025; 16:3553-3574. [PMID: 40230319 DOI: 10.1039/d5fo00475f] [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: 04/16/2025]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome characterized by hypertension, metabolic disorders, and impaired diastolic function, with limited therapeutic options. Recent studies have highlighted the role of ferroptosis in the pathogenesis of HFpEF, and the inhibition of ferroptosis occurrence can significantly improve cardiac function. Limonin, a bioactive ingredient derived from citrus fruits, has been confirmed to exert potential anti-inflammatory and antioxidant effects in some cardiovascular diseases. This study aims to investigate the therapeutic effects of limonin on HFpEF and the underlying mechanisms of inhibiting ferroptosis. HFpEF mice were established by a combination of Nω-nitro-L-arginine methyl ester and a high-fat diet for 6 weeks. Subsequently, the HFpEF mice were treated with empagliflozin or limonin via oral gavage for an additional 6 weeks. Limonin curbed body weight gain and improved metabolic disorders and hypertension. Limonin also ameliorated concentric cardiac hypertrophy and diastolic dysfunction. Transcriptomics and metabolomics analyses revealed that limonin regulated ferroptosis-related pathways and lipid peroxidation. In vivo, limonin improved mitochondrial morphology, reduced cardiac Fe2+ levels and ferroptosis markers such as ROS, 4-HNE and MDA, and increased GSH levels, thereby enhancing antioxidant capacity. Mechanistically, limonin regulated the P53/SLC7A11/GPX4 signaling pathway, promoted the nuclear translocation of Nrf2 (its upstream signaling molecule), and subsequently activated its downstream antioxidant elements, ultimately inhibiting ferroptosis. Furthermore, limonin decreased the expressions of ACSL4, COX2, and ALOXs, which reduced the accumulation of lipid peroxides. These results demonstrate that limonin ameliorates HFpEF by targeting ferroptosis via modulation of the Nrf2/SLC7A11/GPX4 axis, providing a novel strategy for HFpEF treatment.
Collapse
Affiliation(s)
- Xiaoqi Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xinyi Fan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Wangjing Chai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jinling Xiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Jiong Zhao
- Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen 518116, China.
| | - Aolong He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xianwen Tang
- Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen 518116, China.
| | - Fanghe Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Shuzhen Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
16
|
Zhao Y, Qin G, Fan W, Zhang Y, Peng H. TF and TFRC regulate ferroptosis in swine testicular cells through the JNK signaling pathway. Int J Biol Macromol 2025; 307:142369. [PMID: 40120870 DOI: 10.1016/j.ijbiomac.2025.142369] [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: 01/08/2025] [Revised: 02/23/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Transferrin (TF) is a serum glycoprotein that plays a critical role in iron metabolism and typically functions through binding to its transferrin receptor (TFRC). TF is also considered a key indicator of sperm quality and, together with TFRC, plays a critical role in regulating spermatogenesis. This study aimed to explore the effects of increased TF and TFRC expression on ferroptosis in swine testicular cells (ST cells). Our findings revealed that the overexpression of either TF or TFRC diminishes ST cell viability, increases cytotoxicity, intensifies oxidative stress damage, decreases mitochondrial activity, and promotes ferroptosis. Transcriptomic analysis suggested that TF and TFRC may influence ST cells through the MAPK signaling pathway. Subsequent experiments revealed that inhibiting the JNK signaling pathway within the MAPK pathway improved mitochondrial activity, reduced oxidative stress damage, and mitigated ferroptosis progression. Moreover, we discovered that TF and TFRC might regulate cellular oxidative phosphorylation via the JNK signaling pathway. In conclusion, increased expression of TF or TFRC increases the sensitivity of ST cells to ferroptosis and modulates mitochondrial DNA transcription and energy metabolism through the JNK signaling pathway. These findings could offer potential therapeutic targets for addressing reproductive toxicity associated with ferroptosis.
Collapse
Affiliation(s)
- Yuanjie Zhao
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China; College of Life and Health, Hainan University, Haikou 570228, China
| | - Ge Qin
- College of Animal Science and Technology, Southwest University, Chongqing 404100, China
| | - Weiqin Fan
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Yanyan Zhang
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China
| | - Hui Peng
- School of Tropical Agriculture and Forestry, Hainan University, Haikou 570228, China.
| |
Collapse
|
17
|
Barbuti PA, Guardia-Laguarta C, Yun T, Chatila ZK, Flowers X, Wong C, Santos BFR, Larsen SB, Lotti JS, Hattori N, Bradshaw E, Dettmer U, Fanning S, Menon V, Reddy H, Teich AF, Krüger R, Area-Gomez E, Przedborski S. The role of alpha-synuclein in synucleinopathy: Impact on lipid regulation at mitochondria-ER membranes. NPJ Parkinsons Dis 2025; 11:103. [PMID: 40307230 PMCID: PMC12043847 DOI: 10.1038/s41531-025-00960-x] [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: 07/29/2024] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
The protein alpha-synuclein (αSyn) plays a pivotal role in the pathogenesis of synucleinopathies, including Parkinson's disease and multiple system atrophy, with growing evidence indicating that lipid dyshomeostasis is a key phenotype in these neurodegenerative disorders. Previously, we identified that αSyn localizes, at least in part, to mitochondria-associated endoplasmic reticulum membranes (MAMs), which are transient functional domains containing proteins that regulate lipid metabolism, including the de novo synthesis of phosphatidylserine. In the present study, we analyzed the lipid composition of postmortem human samples, focusing on the substantia nigra pars compacta of Parkinson's disease and controls, as well as three less affected brain regions of Parkinson's donors. To further assess synucleinopathy-related lipidome alterations, similar analyses were performed on the striatum of multiple system atrophy cases. Our data reveal region- and disease-specific changes in the levels of lipid species. Specifically, our data revealed alterations in the levels of specific phosphatidylserine species in brain areas most affected in Parkinson's disease. Some of these alterations, albeit to a lesser degree, are also observed in multiple system atrophy. Using induced pluripotent stem cell-derived neurons, we show that αSyn regulates phosphatidylserine metabolism at MAM domains, and that αSyn dosage parallels the perturbation in phosphatidylserine levels. These findings support the notion that αSyn pathophysiology is linked to the dysregulation of lipid homeostasis, which may contribute to the vulnerability of specific brain regions in synucleinopathy. These findings have significant therapeutic implications.
Collapse
Affiliation(s)
- Peter A Barbuti
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Cristina Guardia-Laguarta
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Taekyung Yun
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, Spain
| | - Zena K Chatila
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Xena Flowers
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY, USA
| | - Chantel Wong
- Department of Neuroscience, Barnard College of Columbia University, New York, NY, USA
| | - Bruno F R Santos
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Luxembourg City, Luxembourg
- Disease Modelling and Screening Platform, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - Simone B Larsen
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
| | - James S Lotti
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Elizabeth Bradshaw
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University, New York, NY, USA
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Saranna Fanning
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Vilas Menon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Hasini Reddy
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew F Teich
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Rejko Krüger
- Translational Neuroscience, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belval, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Estela Area-Gomez
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA
- Center for Biological Research (CIB), - Margarita Salas, CSIC, Madrid, Spain
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Serge Przedborski
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
- Center for Motor Neuron Biology and Diseases, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Neuroscience, Columbia University, New York, NY, USA.
| |
Collapse
|
18
|
Zhang W, Li R, Lu D, Wang X, Wang Q, Feng X, Qi S, Zhang X. Phospholipids and peroxisomes in ferroptosis: the therapeutic target of acupuncture regulating vascular cognitive impairment and dementia. Front Aging Neurosci 2025; 17:1512980. [PMID: 40365351 PMCID: PMC12070441 DOI: 10.3389/fnagi.2025.1512980] [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: 10/17/2024] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
Ferroptosis, since its conceptualization in 2012, has witnessed an exponential growth in research interest over recent years. It is regulated by various cellular metabolic pathways during chronic cerebral ischemia and hypoxia, including reactive oxygen species (ROS) generation, iron accumulation, abnormalities in glutathione metabolism, and disruptions in lipid and glucose metabolism. With the deepening and widespread research, ferroptosis has emerged as a critical pathway in the pathogenesis of vascular cognitive impairment and dementia (VCID). This unique cell death pathway caused by iron-dependent phospholipid peroxidation is strongly related to VICD. We examine the impact of phospholipid composition on neuronal susceptibility to ferroptosis, with a particular focus on the critical role of polyunsaturated fatty acids (PUFAs) in this process. Intriguingly, peroxisomes, as key regulators of lipid metabolism and oxidative stress, influence the susceptibility of neuronal cells to ferroptosis through the synthesis of plasmalogens and other lipid species. In this Review, we provide a critical analysis of the current molecular mechanisms and regulatory networks of acupuncture for ferroptosis, the potential functions of acupuncture in peroxisomal functions and phospholipid metabolism, and its neuroprotective effects in VCID, together with a potential for therapeutic targeting. As such, this highlights the theoretical basis for the application of acupuncture in VCID through multi-target regulation of ferroptosis. This review underscores the potential of acupuncture as a non-pharmacological therapeutic approach in VCID, offering new insights into its role in modulating ferroptosis and associated metabolic pathways for neuroprotection.
Collapse
Affiliation(s)
- Wenyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruiyu Li
- Beijing University of Chinese Medicine Shenzhen Hospital (Longgang), Shenzhen, China
| | - Donglei Lu
- Sports Training Academy of Tianjin University of Sport, Tianjin, China
| | - Xinliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiuxuan Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuyang Feng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Sai Qi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuezhu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
19
|
Teng D, Swanson KD, Wang R, Zhuang A, Wu H, Niu Z, Cai L, Avritt FR, Gu L, Asara JM, Zhang Y, Zheng B. DHODH modulates immune evasion of cancer cells via CDP-Choline dependent regulation of phospholipid metabolism and ferroptosis. Nat Commun 2025; 16:3867. [PMID: 40274823 PMCID: PMC12022163 DOI: 10.1038/s41467-025-59307-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
The ability of cancer cells to evade immune destruction is governed by various intrinsic factors including their metabolic state. Here we demonstrate that inactivation of dihydroorotate dehydrogenase (DHODH), a pyrimidine synthesis enzyme, increases cancer cell sensitivity to T cell cytotoxicity through induction of ferroptosis. Lipidomic and metabolomic analyses reveal that DHODH inhibition reduces CDP-choline level and attenuates the synthesis of phosphatidylcholine (PC) via the CDP-choline-dependent Kennedy pathway. To compensate this loss, there is increased synthesis from phosphatidylethanolamine via the phospholipid methylation pathway resulting in increased generation of very long chain polyunsaturated fatty acid-containing PCs. Importantly, inactivation of Dhodh in cancer cells promotes the infiltration of interferon γ-secreting CD8+ T cells and enhances the anti-tumor activity of PD-1 blockade in female mouse models. Our findings reveal the importance of DHODH in regulating immune evasion through a CDP-choline dependent mechanism and implicate DHODH as a promising target to improve the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Da Teng
- Cedars-Sinai Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, USA
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Kenneth D Swanson
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Ruiheng Wang
- Cedars-Sinai Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, USA
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Aojia Zhuang
- Cedars-Sinai Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Haofeng Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Zhixin Niu
- Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Li Cai
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Faith R Avritt
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Lei Gu
- Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Bin Zheng
- Cedars-Sinai Cancer Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, USA.
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA.
| |
Collapse
|
20
|
Hong W, Zeng X, Ma R, Tian Y, Miu H, Ran X, Song R, Luo Z, Ju D, Ma D, Ashrafizadeh M, Bhutia SK, Conde J, Sethi G, Huang H, Duan C. Age-associated reduction in ER-Mitochondrial contacts impairs mitochondrial lipid metabolism and autophagosome formation in the heart. Cell Death Differ 2025:10.1038/s41418-025-01511-w. [PMID: 40254645 DOI: 10.1038/s41418-025-01511-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025] Open
Abstract
The accumulation of dysfunctional giant mitochondria is a hallmark of aged cardiomyocytes. This study investigated the core mechanism underlying this phenomenon, focusing on the disruption of mitochondrial lipid metabolism and its effects on mitochondrial dynamics and autophagy, using both naturally aging mouse models and etoposide-induced cellular senescence models. In aged cardiomyocytes, a reduction in endoplasmic reticulum-mitochondrial (ER-Mito) contacts impairs lipid transport and leads to insufficient synthesis of mitochondrial phosphatidylethanolamine (PE). A deficiency in phosphatidylserine decarboxylase (PISD) further hinders the conversion of phosphatidylserine to PE within mitochondria, exacerbating the deficit of PE production. This PE shortage disrupts autophagosomal membrane formation, leading to impaired autophagic flux and the accumulation of damaged mitochondria. Modulating LACTB expression to enhance PISD activity and PE production helps maintain mitochondrial homeostasis and the integrity of aging cardiomyocytes. These findings highlight the disruption of mitochondrial lipid metabolism as a central mechanism driving the accumulation of dysfunctional giant mitochondria in aged cardiomyocytes and suggest that inhibiting LACTB expression could serve as a potential therapeutic strategy for mitigating cardiac aging and preserving mitochondrial function.
Collapse
Affiliation(s)
- Weilong Hong
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Xue Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, P.R. China
| | - Ruiyan Ma
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, P.R. China
| | - Yu Tian
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Huimin Miu
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Xiaoping Ran
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Rui Song
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Zhenchun Luo
- Intensive Care Unit, Chongqing Traditional Chinese Medicine Hospital, Chongqing, P.R. China
| | - Dapeng Ju
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Daqing Ma
- Perioperative and Systems Medicine Laboratory, Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, P.R. China
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Milad Ashrafizadeh
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, 769008, Odisha, India
| | - João Conde
- Comprehensive Health Research Centre (CHRC), NOVA Medical School, Faculdade de Ciências Médicas, NMS | FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - He Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China.
| | - Chenyang Duan
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China.
| |
Collapse
|
21
|
Jing F, Zhao M, Xiong H, Zeng X, Jiang J, Li T. Mechanisms underlying targeted mitochondrial therapy for programmed cardiac cell death. Front Physiol 2025; 16:1548194. [PMID: 40292006 PMCID: PMC12021874 DOI: 10.3389/fphys.2025.1548194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Heart diseases are common clinical diseases, such as cardiac fibrosis, heart failure, hypertension and arrhythmia. Globally, the incidence rate and mortality of heart diseases are increasing by years. The main mechanism of heart disease is related to the cellular state. Mitochondrion is the organ of cellular energy supply, participating in various signal transduction pathways and playing a vital role in the occurrence and development of heart disease. This review summarizes the cell death patterns and molecular mechanisms associated with heart disease and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Fengting Jing
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Min Zhao
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Hemin Xiong
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Xin Zeng
- School of Continuing Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
22
|
Mishima E, Nakamura T, Doll S, Proneth B, Fedorova M, Pratt DA, Friedmann Angeli JP, Dixon SJ, Wahida A, Conrad M. Recommendations for robust and reproducible research on ferroptosis. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00843-2. [PMID: 40204928 DOI: 10.1038/s41580-025-00843-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 04/11/2025]
Abstract
Ferroptosis is a necrotic, non-apoptotic cell death modality triggered by unrestrained iron-dependent lipid peroxidation. By unveiling the regulatory mechanisms of ferroptosis and its relevance to various diseases, research over the past decade has positioned ferroptosis as a promising therapeutic target. The rapid growth of this research field presents challenges, associated with potentially inadequate experimental approaches that may lead to misinterpretations in the assessment of ferroptosis. Typical examples include assessing whether an observed phenotype is indeed linked to ferroptosis, and selecting appropriate animal models and small-molecule modulators of ferroptotic cell death. This Expert Recommendation outlines state-of-the-art methods and tools to reliably study ferroptosis and increase the reproducibility and robustness of experimental results. We present highly validated compounds and animal models, and discuss their advantages and limitations. Furthermore, we provide an overview of the regulatory mechanisms and the best-studied players in ferroptosis regulation, such as GPX4, FSP1, SLC7A11 and ACSL4, discussing frequent pitfalls in experimental design and relevant guidance. These recommendations are intended for researchers at all levels, including those entering the expanding and exciting field of ferroptosis research.
Collapse
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Sebastian Doll
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Science, University of Ottawa, Ottawa, Ontario, Canada
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany.
- Translational Redox Biology, TUM Natural School of Sciences, Technical University of Munich, Garching, Germany.
| |
Collapse
|
23
|
Lee WC, Dixon SJ. Mechanisms of ferroptosis sensitization and resistance. Dev Cell 2025; 60:982-993. [PMID: 40199240 DOI: 10.1016/j.devcel.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/08/2024] [Accepted: 02/05/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis is an iron-dependent and oxidative form of non-apoptotic cell death with roles in development, homeostasis, and disease. Ferroptosis sensitivity can vary between cells, often for reasons that are not well understood. In this perspective, we describe the core ferroptosis mechanism and outline how changes in iron, redox, and lipid metabolism can alter ferroptosis sensitivity. We propose the concept of a ferroptosis sensitivity-resistance continuum to describe how different intrinsic and extrinsic factors interact to push cells toward a more ferroptosis-sensitive or ferroptosis-resistant state, with effects on development and diseases such as cancer.
Collapse
Affiliation(s)
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
24
|
Wang C, Zhao Y, Peng Y, Chen W, Zhu J, Gu C, Huo R, Ding L, Liu Y, Liu T, Zhang C, Yang W, Wang H, Guo W, Wang B. Juglone induces ferroptotic effect on hepatocellular carcinoma and pan-cancer via the FOSL1-HMOX1 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156417. [PMID: 39923427 DOI: 10.1016/j.phymed.2025.156417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Drug therapy plays an essential role in the management of hepatocellular carcinoma (HCC). Recently, the use of natural products to suppress tumor cells has emerged as a promising direction for drug development. Juglone, a natural compound, exhibits anticancer activities across various cancer types. However, the precise mechanism underlying the anticancer effect of juglone, especially in HCC, remains elusive. PURPOSE This study aimed to investigate the potential inhibitory effects of juglone on HCC and pan-cancer, as well as elucidate the underlying mechanism. METHODS Cell Counting Kit-8 and colony formation assays were used to examine cell proliferation. Transwell and wound healing assays were used to evaluate cell migration. Cell cycle distribution was assessed by flow cytometry. The in vivo effect of juglone on HCC was evaluated by establishing the HCC xenograft mice model. RNA sequencing and inhibitors targeting diverse modes of programmed cell death were applied to uncover the form of juglone-induced cell death. Integrated transcriptomic, and proteomic analyses unveiled the underlying mechanism. The dual-luciferase reporter assay was employed to verify the findings. The pan-cancer value of juglone was assessed using TCGA database analysis and cellular assays. RESULTS Juglone suppressed HCC growth via ferroptosis in vitro and in vivo, which is evidenced by increased levels of iron, lipid peroxidation (LPO), reactive oxygen species (ROS), malondialdehyde (MDA), and decreased levels of glutathione (GSH). Omic analyses, gene silencing and functional analyses showed the upregulated HMOX1 and FOSL1 were the key effector molecule and transcriptional factor in juglone-induced ferroptosis, respectively. The binding site of FOSL1 at the promoter of HMOX1 was identified. Juglone could induce ferroptosis in pan-cancer by activating the FOSL1-HMOX1 axis. CONCLUSION Our findings, for the first time, demonstrate that juglone effectively inhibits tumor growth by inducing FOSL1-HMOX1-dependent ferroptosis, thereby offering a promising strategy for the development of anticancer drugs.
Collapse
Affiliation(s)
- Chuyu Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ying Zhao
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yingfei Peng
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Wei Chen
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Jie Zhu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Chenzheng Gu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ran Huo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Lin Ding
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Yu Liu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Chunyan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China; Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, PR China; Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, PR China; Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, PR China
| | - Wenjing Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Hao Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China; Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, PR China; Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, PR China; Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, PR China.
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, PR China; Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, PR China; Department of Laboratory Medicine, Wusong Central Hospital, Baoshan District, Shanghai, PR China; Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, PR China.
| |
Collapse
|
25
|
Xiao Y, Li Y, Gu J, Lu S, Yu S, Song C. Circadian rhythm gene cryptochrome 2 (Cry2) interacts with lipid metabolism to promote vascular aging. Arch Gerontol Geriatr 2025; 131:105761. [PMID: 39879691 DOI: 10.1016/j.archger.2025.105761] [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: 10/16/2024] [Revised: 12/19/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Vascular aging is the basis of many chronic diseases of the aged, such as hypertension, coronary heart disease and stroke. OBJECTIVE This study aims to deepen our understanding of the pathological mechanisms of vascular aging by combining multiple big data research methods, and reveal potential therapeutic targets and biomarkers. METHODS WGCNA method was used to integrate the aortic transcriptome data of multiple age stages, and extract the key module and key pathway. The gene of aortic rhythm was integrated by JTK algorithm. Correlation calculation was performed for core gene and associated pathways. Finally, the expression of the core gene and their interaction with the associated pathways were verified in cell senescence. RESULTS WGCNA showed that circadian rhythm is the key pathway of vascular aging, and circadian rhythm and metabolism interact to promote the occurrence of vascular aging. Cry2 has been identified as the most critical core rhythm gene. Lipid metabolism is the most Cry2-related subpathway, among which phospholipid metabolism and Serac1 have the strongest and most significant correlation with Cry2. Cry2 is mainly distributed in endothelial cells in both young and senescent blood vessels, and affects five lipid-related metabolic processes including lipid transport during endothelial senescence. CONCLUSION This study suggests that circadian rhythm and Cry2 may be potential targets of vascular aging, and further studies on their interaction with lipid metabolism will provide effective strategies for the prevention and treatment of age-related vascular diseases.
Collapse
Affiliation(s)
- Yu Xiao
- Department of special needs ward and general practice, Second Affiliated Hospital of Jilin University, Changchun 130041, PR China
| | - Yang Li
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun 130041, PR China
| | - Jinning Gu
- Department of special needs ward and general practice, Second Affiliated Hospital of Jilin University, Changchun 130041, PR China
| | - Shan Lu
- Department of special needs ward and general practice, Second Affiliated Hospital of Jilin University, Changchun 130041, PR China
| | - Shuang Yu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Jilin University, Changchun 130041, PR China
| | - Chunli Song
- Department of special needs ward and general practice, Second Affiliated Hospital of Jilin University, Changchun 130041, PR China.
| |
Collapse
|
26
|
Wu Y, Li C, Lu D, Chen K, Su R, Xu S, Gao F, Lian Z, Yang F, Chen J, Wei F, Xu X, Liu Z. Insulin-induced gene 2 alleviates ischemia-reperfusion injury in steatotic liver by inhibiting GPX4-dependent ferroptosis. Cell Death Discov 2025; 11:127. [PMID: 40169542 PMCID: PMC11962074 DOI: 10.1038/s41420-025-02406-y] [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: 10/14/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/03/2025] Open
Abstract
Hepatic steatosis significantly elevates the vulnerability of the graft to ischemia-reperfusion (I/R) injury during liver transplantation (LT). We investigated the protective role of insulin-induced gene 2 (Insig2) in steatotic liver's I/R injury and underlying mechanisms. Employing mouse model with Insig2 knock-out or hepatocyte-specific overexpression and high-fat diets to induce steatosis, we subjected these mice to hepatic I/R injury. The primary hepatocytes isolated from steatotic liver were used in in vitro hypoxia/reoxygenation (H/R) experiment. Our integrated in vivo and in vitro approach uncovered that Insig2 deficiency exacerbated steatotic liver's damage following hepatic I/R injury, whereas its overexpression offers protection. Mechanically, integrative analysis of transcriptome, proteome, and metabolome found that Insig2 deficiency disturbed lipid metabolism and oxidative stress homeostasis, particularly inhibiting GPX4 expression to induce ferroptosis. Furthermore, chemical inhibition of ferroptosis reversed the deleterious effect of Insig2 deficiency; whereas the protective influence of Insig2 overexpression was negated by the target inhibition of GPX4, leading to an exacerbation of hepatic I/R damage. These insights underscored the potential of the Insig2-GPX4 axis as a therapeutic target, presenting a novel avenue for enhancing the resilience of steatotic liver grafts against I/R injury.
Collapse
Affiliation(s)
- Yichao Wu
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Changbiao Li
- Department of Gastrointestinal-Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Di Lu
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Institution of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Kangchen Chen
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou, China
| | - Renyi Su
- Zhejiang University School of Medicine, Hangzhou, China
| | - Shengjun Xu
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou, China
| | - Fengqiang Gao
- Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengxing Lian
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Fan Yang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Hangzhou, China
| | - Jun Chen
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Institution of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Fangqiang Wei
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
- Institution of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Xiao Xu
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.
- Institution of Clinical Medicine, Hangzhou Medical College, Hangzhou, China.
| | - Zhikun Liu
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.
- Institution of Clinical Medicine, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
27
|
Sun P, Liu Q, Yuan S, Wang XT, Qiu Y, Ge XY. SARS-CoV-2 Membrane Protein Induces MARCHF1/GPX4-Mediated Ferroptosis by Promoting Lipid Accumulation. J Med Virol 2025; 97:e70328. [PMID: 40186530 DOI: 10.1002/jmv.70328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 02/21/2025] [Accepted: 03/14/2025] [Indexed: 04/07/2025]
Abstract
The membrane protein (M), a key structural protein of SARS-CoV-2 that regulates virus assembly and morphogenesis, is involved in the pathological processes of multiple organ damage and metabolic disorders. This study aims to elucidate the mechanisms of M-mediated host ferroptosis and lipid accumulation during SARS-CoV-2 infection. Here, we detected that M protein enhances cellular sensitivity to ferroptosis. Additionally, we uncovered the pivotal role of perilipin-2 and sterol regulatory element-binding protein 1 in M-induced lipid accumulation. Xanthohumol, a cost-effective and orally available diacylglycerol acyltransferase inhibitor, alleviated triglyceride and total cholesterol accumulation, thereby counteracting the M-induced ferroptosis. Furthermore, we identified that the mitochondrial import inner membrane translocase subunit TIM23 and the mitochondrial import receptor subunit TOM20 homolog contribute to M-induced mitochondrial dysfunction. Notably, inhibiting lipid synthesis effectively reduced mitochondrial reactive oxygen species and transmembrane potential, indicating a cross-talk between lipid and ferro metabolic pathways. Mechanistically, glutathione peroxidase 4 (GPX4) interacts with SARS-CoV-2 M, leading to its subsequent degradation by the Membrane Associated Ring-CH-Type Finger 1 (MARCHF1) ubiquitin ligase. M-GPX4 interaction occurs at the R72 residue, which may represent a potential therapeutic target against SARS-CoV-2 infection. M modulates lipid accumulation and further impairs mitochondrial functions, ultimately resulting in ferroptosis through MARCHF1-GPX4 axis. Disrupting host-virus interactions along this pathway may provide a therapeutic strategy for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Pei Sun
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Qian Liu
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Shuofeng Yuan
- Department of Microbiology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xin-Tao Wang
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Ye Qiu
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Xing-Yi Ge
- Department of Biomedical Engineering, Hunan Provincial Key Laboratory of Medical Virology, College of Biology, Hunan University, Changsha, Hunan, China
| |
Collapse
|
28
|
Zheng J, Conrad M. Ferroptosis: when metabolism meets cell death. Physiol Rev 2025; 105:651-706. [PMID: 39661331 DOI: 10.1152/physrev.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
We present here a comprehensive update on recent advancements in the field of ferroptosis, with a particular emphasis on its metabolic underpinnings and physiological impacts. After briefly introducing landmark studies that have helped to shape the concept of ferroptosis as a distinct form of cell death, we critically evaluate the key metabolic determinants involved in its regulation. These include the metabolism of essential trace elements such as selenium and iron; amino acids such as cyst(e)ine, methionine, glutamine/glutamate, and tryptophan; and carbohydrates, covering glycolysis, the citric acid cycle, the electron transport chain, and the pentose phosphate pathway. We also delve into the mevalonate pathway and subsequent cholesterol biosynthesis, including intermediate metabolites like dimethylallyl pyrophosphate, squalene, coenzyme Q (CoQ), vitamin K, and 7-dehydrocholesterol, as well as fatty acid and phospholipid metabolism, including the biosynthesis and remodeling of ester and ether phospholipids and lipid peroxidation. Next, we highlight major ferroptosis surveillance systems, specifically the cyst(e)ine/glutathione/glutathione peroxidase 4 axis, the NAD(P)H/ferroptosis suppressor protein 1/CoQ/vitamin K system, and the guanosine triphosphate cyclohydrolase 1/tetrahydrobiopterin/dihydrofolate reductase axis. We also discuss other potential anti- and proferroptotic systems, including glutathione S-transferase P1, peroxiredoxin 6, dihydroorotate dehydrogenase, glycerol-3-phosphate dehydrogenase 2, vitamin K epoxide reductase complex subunit 1 like 1, nitric oxide, and acyl-CoA synthetase long-chain family member 4. Finally, we explore ferroptosis's physiological roles in aging, tumor suppression, and infection control, its pathological implications in tissue ischemia-reperfusion injury and neurodegeneration, and its potential therapeutic applications in cancer treatment. Existing drugs and compounds that may regulate ferroptosis in vivo are enumerated.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
- Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, Garching, Germany
| |
Collapse
|
29
|
Chen Z, Wang Y, Li Z, Chen M, Li Y, Lu C, Lin Z, Zheng H, Chen L, Zhang Q. Improving ferroptosis-mediated immunotherapy for colorectal cancer through lysosome-targeted photodynamic therapy. Mater Today Bio 2025; 31:101552. [PMID: 40018057 PMCID: PMC11867524 DOI: 10.1016/j.mtbio.2025.101552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/10/2025] [Accepted: 02/03/2025] [Indexed: 03/01/2025] Open
Abstract
Lysosomes is emerging as a promising therapeutic target for improving immunotherapy, which dysfunction would trigger lysosomal membrane permeabilization increase and subsequent leakage of reduced iron, which contributed to ferroptosis through cell-intrinsic Fenton chemistry. However, the integrity of lysosomal membranes is not susceptible to disrupt, owing to the presence of several Endo-lysosomal damage-response mechanisms. Herein, we developed a lysosome-targeted photosensitizer (TLA), which possessed robust light stability, good bio-compatibility, and high photodynamic therapy (PDT) effect. Upon internalized by cancer cells, TLA was specifically accumulated in lysosome, and which would destroy the integrity of lysosomal membranes and inhibit protective autophagy upon exposure to light irradiation. Subsequently, the cancer cells were suffered from ferroptosis through triggering cell-intrinsic Fenton chemistry and mitochondrial dysfunction, which would release damage-associated molecular pattern molecules (DAMPs) to induce immunogenic cell death and remodel immunosuppressive tumor microenvironment. Notably, combined with PD-L1 antibody and TLA could greatly potentiate the immune response and exhibit highest anti-tumor effects. In summary, this novel lysosome-targeted photosensitizer could serve as a promising strategy for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Zhian Chen
- Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Yutong Wang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhenhao Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Meijuan Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Hepatology Unit and Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yingshi Li
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chuyue Lu
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhenyu Lin
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hua Zheng
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lujia Chen
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianbing Zhang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
30
|
Liu S, Ma Z. The role of cannabinoid-mediated signaling pathways and mechanisms in brain disorders. Cell Signal 2025; 128:111653. [PMID: 39952540 DOI: 10.1016/j.cellsig.2025.111653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/17/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Cannabinoids play significant roles in the central nervous system (CNS), but cannabinoid-mediated physiopathological functions are not elaborated. Cannabinoid receptors (CBRs) mediate functions that include the regulation of neuroinflammation, oxidative stress, apoptosis, autophagy, and neurogenesis. Microglia are the primary immune cells responsible for mediating neuroinflammation in the CNS. Therefore, this article primarily focuses on microglia to summarize the inflammatory pathways mediated by cannabinoids in the CNS, including nuclear factor-κB (NF-κB), NOD-like receptor protein 3 (NLRP3) inflammasome, mitogen-activated protein kinase (MAPK), protein kinase B (Akt), and cAMP-dependent protein kinase (PKA) signaling pathways. Additionally, we provide a table summarizing the role of cannabinoids in various brain diseases. Medical use of cannabinoids has protective effects in preventing and treating brain diseases; however, excessive and repeated use can be detrimental to the CNS. We propose that cannabinoids hold significant potential for preventing and treating brain diseases, including ferroptosis, lactate metabolism, and mitophagy, providing new insights for further research on cannabinoids.
Collapse
Affiliation(s)
- Shunfeng Liu
- School of Basic Medicine, Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disorders, Qingdao University, Qingdao 266071, China
| | - Zegang Ma
- School of Basic Medicine, Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disorders, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
31
|
Liu Y, Qiu S, Huang H, Wu Z, Ge S. Ferrostatin supplementation improves microalgal activities and nutrient removal in wastewater under high temperature shock: From ferroptosis-like inhibition to enhanced oxidation resistance. WATER RESEARCH 2025; 273:123033. [PMID: 39721506 DOI: 10.1016/j.watres.2024.123033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
High temperature (HT) shock is one of environmental stressors suppressing microalgal activities in microalgal wastewater bioremediation system. However, its inhibition mechanism and how to alleviate such suppression remain inadequately understood. This study confirmed a transient ferroptosis as a novel form of programmed cell death in a wastewater-indigenous Chlorella sp., responding to a 30-minute HT (50 °C) exposure, through the systematically physiological, metabolomic and transcriptomic analysis. Specifically, the HT-induced ferroptosis could be supported by both the growth and physiological indicators. These include the suppressed growth (76.05 %), suppressed nutrient removals (NH4+-N by 76.22 %, PO43--P by 64.15 %), accumulated intracellular Fe3+ concentrations (7.75-fold), enhanced oxidative stress (e.g., increased levels of reactive oxygen species (159.97 %)), activated antioxidant defense system (e.g., increased activities of superoxide dismutase (24.83 %) and catalase (5.03-fold)), and obvious membrane damage (e.g., increased levels of malondialdehyde (1.67-fold)). Further metabolomic analysis indicated that such HT-induced ferroptosis was also largely related to the significant alternations of lipid remodeling in three aspects: varied abundance of certain lipids specific to chloroplast membrane or mitochondria, accumulation of certain lipids with lower unsaturation, and formation of lipid peroxides disrupting membrane integrity. Moreover, the key genes involved in ferroptosis correspondingly responded, especially those associated with lipid metabolism (e.g., ACSL), antioxidant defense system (e.g., GSS, GPX and GSR), mitochondrial normal functioning (e.g., SEL1L), autophagy regulation (e.g., ATG9, ATG11, ATG13) and protein folding (e.g., HSPA5, HSPA1s, HSP90B). In addition, the supplementation of the typical ferroptosis inhibitor Ferrostatin-1 effectively mitigated lipid peroxide accumulation and suppressed the onset of ferroptosis, accelerating subsequent recovery of NH4+-N removal by 60.66 %. These findings update current understandings of microalgal ferroptosis-like inhibition, offering Ferrostatin-1 supplementation as a potential strategy for system resistance to heat stress in microalgae-based bioremediation system.
Collapse
Affiliation(s)
- Yanlin Liu
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China
| | - Shuang Qiu
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China
| | - Hongtao Huang
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China
| | - Zhengshuai Wu
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China
| | - Shijian Ge
- Jiangsu Key Laboratory of Chemical Pollution Control and Resources Reuse, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Xiao Ling Wei 200, Nanjing 210094, Jiangsu, China.
| |
Collapse
|
32
|
Selvaraj NR, Nandan D, Nair BG, Nair VA, Venugopal P, Aradhya R. Oxidative Stress and Redox Imbalance: Common Mechanisms in Cancer Stem Cells and Neurodegenerative Diseases. Cells 2025; 14:511. [PMID: 40214466 PMCID: PMC11988017 DOI: 10.3390/cells14070511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/14/2025] Open
Abstract
Oxidative stress (OS) is an established hallmark of cancer and neurodegenerative disorders (NDDs), which contributes to genomic instability and neuronal loss. This review explores the contrasting role of OS in cancer stem cells (CSCs) and NDDs. Elevated levels of reactive oxygen species (ROS) contribute to genomic instability and promote tumor initiation and progression in CSCs, while in NDDs such as Alzheimer's and Parkinson's disease, OS accelerates neuronal death and impairs cellular repair mechanisms. Both scenarios involve disruption of the delicate balance between pro-oxidant and antioxidant systems, which leads to chronic oxidative stress. Notably, CSCs and neurons display alterations in redox-sensitive signaling pathways, including Nrf2 and NF-κB, which influence cell survival, proliferation, and differentiation. Mitochondrial dynamics further illustrate these differences: enhanced function in CSCs supports adaptability and survival, whereas impairments in neurons heighten vulnerability. Understanding these common mechanisms of OS-induced redox imbalance may provide insights for developing interventions, addressing aging hallmarks, and potentially mitigating or preventing both cancer and NDDs.
Collapse
Affiliation(s)
| | | | | | | | - Parvathy Venugopal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| | - Rajaguru Aradhya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| |
Collapse
|
33
|
Li C, Deng D, Jiang Q, Shi J, Xu L, Liu Y. Ferroptosis in NAFLD: insights and the therapeutic potential of exercise. Front Med (Lausanne) 2025; 12:1462145. [PMID: 40206477 PMCID: PMC11979233 DOI: 10.3389/fmed.2025.1462145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Ferroptosis, a distinct form of non-apoptotic cell death driven by iron accumulation, has garnered significant attention in recent years. Emerging evidence suggests that ferroptosis in hepatocytes may serve as a pivotal trigger in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Importantly, inhibiting ferroptosis has shown promising potential in slowing the progression of NAFLD. Concurrently, exercise, a cornerstone in the prevention and management of chronic diseases, plays a critical role in regulating disease progression. As such, the modulation of ferroptosis through exercise represents a promising avenue for developing innovative therapeutic strategies. This review aims to systematically elucidate the conceptual framework and molecular mechanisms underlying ferroptosis, with particular emphasis on its pathophysiological role in NAFLD. We have systematically summarized the effects of exercise on ferroptosis regulation through multiple molecular mechanisms, including upregulation of antioxidant defense systems via activation of NRF2, GPX4, and SLC7A11 signaling pathways; and modulation of iron metabolism through FPN-mediated iron homeostasis regulation. These findings not only provide valuable insights into the molecular basis of exercise-induced protection against ferroptosis-mediated cellular damage but also offer novel perspectives for future investigations into exercise-based interventions for NAFLD management. This work thereby contributes to the advancement of therapeutic strategies in the field of metabolic liver diseases.
Collapse
Affiliation(s)
- Chang Li
- Graduate School, Harbin Sport University, Harbin, Heilongjiang, China
| | - Dongkun Deng
- Graduate School, Harbin Sport University, Harbin, Heilongjiang, China
| | - Qingfeng Jiang
- Graduate School, Harbin Sport University, Harbin, Heilongjiang, China
| | - Jiaming Shi
- Graduate School, Harbin Sport University, Harbin, Heilongjiang, China
| | - Lin Xu
- College of Human Sport Science, Harbin Sport University, Harbin, Heilongjiang, China
| | - Yufei Liu
- College of Human Sport Science, Harbin Sport University, Harbin, Heilongjiang, China
| |
Collapse
|
34
|
Zhao Y, Zhang J, Xia J, Han X, Ben S, Liu T, Mu W, Yao M, Jiang Q, Yan B. Identification of age-related metabolomic signatures in vascular tissues. Biochem Biophys Res Commun 2025; 754:151513. [PMID: 40010140 DOI: 10.1016/j.bbrc.2025.151513] [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: 11/23/2024] [Revised: 02/04/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Vascular aging contributes to the morbidity and mortality in older individuals, closely linked to an imbalance between energy consumption and production. Despite its importance, our understanding of how aging affects vascular metabolism and leads to vascular diseases remains limited. In this study, we explored the metabolomic characteristics of vascular aging by analyzing aortic tissues from young and old mice through untargeted metabolomic analysis using ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). We identified 85 differential metabolites, with 37 up-regulated and 48 down-regulated, primarily consisting of lipids and lipid-like molecules, based on the criteria of variable importance in projection (VIP) > 1 and P < 0.05. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed significant involvement of these metabolites in six metabolic pathways (P < 0.05), particularly in glycerophospholipid metabolism. Receiver operating characteristic (ROC) curve analysis highlighted eight altered metabolites in glycerophospholipid metabolism, such as phosphatidylcholine (PC) (17:0/22:6) and lysophosphatidylcholine (LPC) (18:2), which demonstrated strong discriminatory ability for vascular aging with an area under the curve (AUC) exceeding 0.85. This study provides novel insights into metabolomic signature of vascular aging, offering important clues for future treatments of age-related vascular disorders.
Collapse
Affiliation(s)
- Ya Zhao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jingyue Zhang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Jiao Xia
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiaoyan Han
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200030, China
| | - Shuai Ben
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Tianyi Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Nantong University, Nantong, 226000, China
| | - Wan Mu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200030, China
| | - Mudi Yao
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China.
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
35
|
Xu SY, Yin SS, Wang L, Zhong H, Wang H, Yu HY. Insights into emerging mechanisms of ferroptosis: new regulators for cancer therapeutics. Cell Biol Toxicol 2025; 41:63. [PMID: 40131564 PMCID: PMC11937073 DOI: 10.1007/s10565-025-10010-0] [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: 09/07/2024] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
Ferroptosis is an iron-dependent form of regulated cell death characterized by the accumulation of iron-dependent lipid peroxides, which has been implicated in the pathogenesis of various diseases, and therapeutic agents targeting ferroptosis are emerging as promising tools for cancer treatment. Current research reveals that ferroptosis-targeted therapies can effectively inhibit tumor progression or delay cancer development. Notably, natural product-derived compounds-such as artemisinin, baicalin, puerarin, quercetin, kaempferol, and apigenin-have demonstrated the ability to modulate ferroptosis, offering potential anti-cancer benefits. Mechanistically, ferroptosis exhibits negative glutathione peroxidase 4 (GPX4) regulation and demonstrates a positive correlation with plasma membrane polyunsaturated fatty acid (PUFA) abundance. Moreover, the labile iron pool (LIP) serves as the redox engine of ferroptosis. This review systematically analyzes the hallmarks, signaling pathways, and molecular mechanisms of ferroptosis, with a focus on how natural product-derived small molecules regulate this process. It further evaluates their potential as ferroptosis inducers or inhibitors in anti-tumor therapy, providing a foundation for future clinical translation.
Collapse
Affiliation(s)
- Si-Yi Xu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shuang-Shuang Yin
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Lei Wang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Hao Zhong
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Hong Wang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Hai-Yang Yu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
36
|
Yao S, Quan Y. Research progress of ferroptosis pathway and its related molecular ubiquitination modification in liver cancer. Front Oncol 2025; 15:1502673. [PMID: 40190567 PMCID: PMC11968660 DOI: 10.3389/fonc.2025.1502673] [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: 09/27/2024] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
As a new type of programmed cell death, ferroptosis is characterized by iron metabolism disorder and reactive oxygen species (ROS) accumulation, and is involved in regulating the occurrence and development of cancer cells. Especially in the field of liver cancer treatment, ferroptosis shows great potential because it can induce tumor cell death. Ubiquitination is a process of protein post-translational modification, which can affect the stability of proteins and regulate the progress of ferroptosis. This article reviews the research progress of ubiquitination modification of molecules related to ferroptosis pathway in the regulation of liver cancer, providing a new strategy for the treatment of liver cancer.
Collapse
Affiliation(s)
- Silin Yao
- The First Clinical Medical School, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yi Quan
- The First People’s Hospital of Zhaoqing, Guangdong Medical University, Zhaoqing, Guangdong, China
| |
Collapse
|
37
|
Sokol KH, Lee CJ, Rogers TJ, Waldhart A, Ellis AE, Madireddy S, Daniels SR, House RRJ, Ye X, Olesnavich M, Johnson A, Furness BR, Sheldon RD, Lien EC. Lipid availability influences ferroptosis sensitivity in cancer cells by regulating polyunsaturated fatty acid trafficking. Cell Chem Biol 2025; 32:408-422.e6. [PMID: 39442523 PMCID: PMC11928283 DOI: 10.1016/j.chembiol.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/09/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Ferroptosis is a form of cell death caused by lipid peroxidation that is emerging as a target for cancer therapy, highlighting the need to identify factors that govern ferroptosis susceptibility. Lipid peroxidation occurs primarily on phospholipids containing polyunsaturated fatty acids (PUFAs). Here, we show that even though extracellular lipid limitation reduces cellular PUFA levels, lipid-starved cancer cells are paradoxically more sensitive to ferroptosis. Using mass spectrometry-based lipidomics with stable isotope fatty acid labeling, we show that lipid limitation induces a fatty acid trafficking pathway in which PUFAs are liberated from triglycerides to synthesize highly unsaturated PUFAs such as arachidonic and adrenic acid. These PUFAs then accumulate in phospholipids, including ether phospholipids, to promote ferroptosis sensitivity. Therefore, PUFA levels within cancer cells do not necessarily correlate with ferroptosis susceptibility. Rather, how cancer cells respond to extracellular lipid levels by trafficking PUFAs into proper phospholipid pools contributes to their sensitivity to ferroptosis.
Collapse
Affiliation(s)
- Kelly H Sokol
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Cameron J Lee
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Thomas J Rogers
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Althea Waldhart
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Abigail E Ellis
- Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Sahithi Madireddy
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Samuel R Daniels
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA; Van Andel Institute Graduate School, Grand Rapids, MI 49503, USA
| | - Rachel Rae J House
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Xinyu Ye
- Van Andel Institute Graduate School, Grand Rapids, MI 49503, USA
| | - Mary Olesnavich
- Van Andel Institute Graduate School, Grand Rapids, MI 49503, USA
| | - Amy Johnson
- Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Benjamin R Furness
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ryan D Sheldon
- Mass Spectrometry Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Evan C Lien
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
38
|
Su Y, Yuan Q. Mitochondrial fatty acid oxidase CPT1A ameliorates postoperative cognitive dysfunction by regulating astrocyte ferroptosis. Brain Res 2025; 1850:149424. [PMID: 39725377 DOI: 10.1016/j.brainres.2024.149424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a significant surgery-related complication marked by cognitive decline. Studies indicated that neuroinflammation, ferroptosis, and mitochondrial fatty acid metabolism might play parts in POCD, and might be mediated by Carnitine palmitoyl transferase 1a (CPT1A), but requires further investigations. Therefore, this study aims to investigate the mechanism of mitochondrial fatty acid oxidase CPT1A on mitochondrial function, ferroptosis, and inflammation in POCD pathogenesis. METHODS SVG P12 astrocytes were used to investigate CPT1A's control over mitochondrial function, ferroptosis, and inflammation affecting neurons. CPT1A was overexpressed using shRNA, with or without oligomycin to modulate mitochondrial function. Co-culture of these astrocytes with neurons, under similar conditions, assessed CPT1A's impact on neuron damage via ferroptosis and inflammation. Gene and protein expressions of CPT1A, SYN, PSD95 were measured via RT-PCR and WB. Detection of JC-1, mitochondrial oxygen consumption rate (OCR), ROS, Fe2+ concentration, MOD, SOD and GSH/GSSG using kits was conducted to explore mitochondrial function and ferroptosis. Inflammation was quantified by ELISA for IL-6, IL-1β, and TGF-β. RESULTS We successfully established CPT1A overexpression and knockdown models in astrocytes, confirming CPT1A's ability to enhance mitochondrial membrane potential. Elevated CPT1A levels led to improved mitochondrial function, synaptic integrity, reduced oxidative stress, maintained iron homeostasis, and attenuated neuroinflammation, as reflected by increased SYN, PSD95, OCR, GSH and SOD, decreased ROS,GSSG, MDA, iron levels, and lowered inflammatory factors expression. Treatment with oligomycin reversed these protective effects, demonstrating the dependency of CPT1A's benefits on intact mitochondrial respiration. In co-culture experiments with hippocampal neurons, astrocytes with manipulated CPT1A levels, particularly those co-treated with oligomycin, exacerbated neuronal mitochondrial dysfunction, oxidative stress, iron accumulation, and inflammation. CONCLUSION Overexpression of mitochondrial fatty acid oxidase CPT1A might improve synaptic integrity and rescue POCD by ameliorating astrocyte ferroptosis and neuroinflammation.
Collapse
Affiliation(s)
- Yinglan Su
- Department of Anesthesiology, Shenzhen University General Hospital, Shenzhen City, Guangdong Province, China.
| | - Qian Yuan
- Department of Urology Surgery, People's Hospital of Shenzhen, Shenzhen City, Guangdong Province, China
| |
Collapse
|
39
|
Pei Z, Fan J, Tang M, Li Y. Ferroptosis: A New Strategy for the Treatment of Fibrotic Diseases. Adv Biol (Weinh) 2025; 9:e2400383. [PMID: 39377183 DOI: 10.1002/adbi.202400383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/07/2024] [Indexed: 10/09/2024]
Abstract
Ferroptosis is a new type of cell death characterized by iron dependence and the excessive accumulation of lipid reactive oxygen species (lipid ROS) that has gradually become better characterized. There is sufficient evidence indicating that ferroptosis is associated with a variety of human life activities and diseases, such as tumor suppression, ischemic organ injury, and degenerative disorders. Notably, ferroptosis is also involved in the initiation and development of fibrosis in various organs, including liver fibrosis, pulmonary fibrosis, renal fibrosis, and cardiac fibrosis, which is usually irreversible and refractory. Although a large number of patients with fibrosis urgently need to be treated, the current treatment options are still limited and unsatisfactory. Organ fibrosis involves a series of complex and orderly processes, such as parenchymal cell damage, recruitment of inflammatory cells and activation of fibroblasts, which ultimately leads to the accumulation of extracellular matrix (ECM) and the formation of fibrosis. An increasing number of studies have confirmed the close association between these pathological processes and ferroptosis. This review summarizes the role and function of ferroptosis in fibrosis and proposes several potential therapeutic strategies and pathways based on ferroptosis.
Collapse
Affiliation(s)
- Zhuo Pei
- Air Force Hospital of the Central Theater Command of PLA, Datong, 037006, China
| | - Jing Fan
- Air Force Hospital of the Northern Theater Command of the People's Liberation Army of China, Shenyang, 110044, China
| | - Maolin Tang
- Air Force Hospital of the Central Theater Command of PLA, Datong, 037006, China
| | - Yuhong Li
- Department of Cell Biology, Army Medical University, Chongqing, 400038, China
| |
Collapse
|
40
|
Zhang Y, Zhang Y, Gong R, Liu X, Zhang Y, Sun L, Ma Q, Wang J, Lei K, Ren L, Zhao C, Zheng X, Xu J, Ren H. Label-Free Prediction of Tumor Metastatic Potential via Ramanome. SMALL METHODS 2025; 9:e2400861. [PMID: 39558758 DOI: 10.1002/smtd.202400861] [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: 06/08/2024] [Revised: 09/02/2024] [Indexed: 11/20/2024]
Abstract
Assessing metastatic potential is crucial for cancer treatment strategies. However, current methods are time-consuming, labor-intensive, and have limited sample accessibility. Therefore, this study aims to investigate the urgent need for rapid and accurate approaches by proposing a Ramanome-based metastasis index (RMI) using machine learning of single-cell Raman spectra to rapidly and accurately assess tumor cell metastatic potential. Validation with various cultured tumor cells and a mouse orthotopic model of pancreatic ductal adenocarcinoma show a Kendall rank correlation coefficient of 1 compared to Transwell experiments and histopathological assessments. Significantly, lipid-related Raman peaks are most influential in determining RMI. The lipidomic analysis confirmed strong correlations between metastatic potential and phosphatidylcholine, phosphatidylethanolamine, cholesteryl ester, ceramide, and bis(monoacylglycero)phosphate, crucial in cell membrane composition or signal transduction. Therefore, RMI is a valuable tool for predicting tumor metastatic potential and providing insights into metastasis mechanisms.
Collapse
Affiliation(s)
- Yuxing Zhang
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yanmei Zhang
- CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - Ruining Gong
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Xiaolan Liu
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yu Zhang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Luyang Sun
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - Qingyue Ma
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Jia Wang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Ke Lei
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Linlin Ren
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Chenyang Zhao
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| | - Xiaoshan Zheng
- CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - Jian Xu
- CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
- Shandong Energy Institute, Qingdao, Shandong, 266101, China
| | - He Ren
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266000, China
| |
Collapse
|
41
|
Gollowitzer A, Pein H, Rao Z, Waltl L, Bereuter L, Loeser K, Meyer T, Jafari V, Witt F, Winkler R, Su F, Große S, Thürmer M, Grander J, Hotze M, Harder S, Espada L, Magnutzki A, Gstir R, Weinigel C, Rummler S, Bonn G, Pachmayr J, Ermolaeva M, Harayama T, Schlüter H, Kosan C, Heller R, Thedieck K, Schmitt M, Shimizu T, Popp J, Shindou H, Kwiatkowski M, Koeberle A. Attenuated growth factor signaling during cell death initiation sensitizes membranes towards peroxidation. Nat Commun 2025; 16:1774. [PMID: 40000627 PMCID: PMC11861335 DOI: 10.1038/s41467-025-56711-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Cell death programs such as apoptosis and ferroptosis are associated with aberrant redox homeostasis linked to lipid metabolism and membrane function. Evidence for cross-talk between these programs is emerging. Here, we show that cytotoxic stress channels polyunsaturated fatty acids via lysophospholipid acyltransferase 12 into phospholipids that become susceptible to peroxidation under additional redox stress. This reprogramming is associated with altered acyl-CoA synthetase isoenzyme expression and caused by a decrease in growth factor receptor tyrosine kinase (RTK)-phosphatidylinositol-3-kinase signaling, resulting in suppressed fatty acid biosynthesis, for specific stressors via impaired Akt-SREBP1 activation. The reduced availability of de novo synthesized fatty acids favors the channeling of polyunsaturated fatty acids into phospholipids. Growth factor withdrawal by serum starvation mimics this phenotype, whereas RTK ligands counteract it. We conclude that attenuated RTK signaling during cell death initiation increases cells' susceptibility to oxidative membrane damage at the interface of apoptosis and alternative cell death programs.
Collapse
Affiliation(s)
- André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Helmut Pein
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Zhigang Rao
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Lorenz Waltl
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Leonhard Bereuter
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Konstantin Loeser
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Tobias Meyer
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Vajiheh Jafari
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Finja Witt
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - René Winkler
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
- Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916, Badalona, Spain
| | - Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Silke Große
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Maria Thürmer
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Julia Grander
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Madlen Hotze
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Sönke Harder
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Lilia Espada
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Alexander Magnutzki
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Ronald Gstir
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Christina Weinigel
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Silke Rummler
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Günther Bonn
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Johanna Pachmayr
- Institute of Pharmacy, Paracelsus Medical University, 5020, Salzburg, Austria
| | - Maria Ermolaeva
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Takeshi Harayama
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur - CNRS UMR7275 - Inserm U1323, 06560, Valbonne, France
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
| | - Regine Heller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Kathrin Thedieck
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
- Department Metabolism, Senescence and Autophagy, Research Center One Health Ruhr, University Alliance Ruhr & University Hospital Essen, University Duisburg-Essen, 45141, Essen, Germany
- Freiburg Materials Research Center FMF, Albert-Ludwigs-University of Freiburg, 79104, Freiburg, Germany
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
- German Cancer Consortium (DKTK), partner site Essen/Duesseldorf, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147, Essen, Germany
| | - Michael Schmitt
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Institute of Microbial Chemistry, Tokyo 141-0021, Japan
| | - Jürgen Popp
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Hideo Shindou
- Department of Lipid Life Science, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Marcel Kwiatkowski
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria.
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany.
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria.
| |
Collapse
|
42
|
Fernández-Acosta R, Vintea I, Koeken I, Hassannia B, Vanden Berghe T. Harnessing ferroptosis for precision oncology: challenges and prospects. BMC Biol 2025; 23:57. [PMID: 39988655 PMCID: PMC11849278 DOI: 10.1186/s12915-025-02154-6] [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: 11/28/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
The discovery of diverse molecular mechanisms of regulated cell death has opened new avenues for cancer therapy. Ferroptosis, a unique form of cell death driven by iron-catalyzed peroxidation of membrane phospholipids, holds particular promise for targeting resistant cancer types. This review critically examines current literature on ferroptosis, focusing on its defining features and therapeutic potential. We discuss how molecular profiling of tumors and liquid biopsies can generate extensive multi-omics datasets, which can be leveraged through machine learning-based analytical approaches for patient stratification. Addressing these challenges is essential for advancing the clinical integration of ferroptosis-driven treatments in cancer care.
Collapse
Affiliation(s)
- Roberto Fernández-Acosta
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Iuliana Vintea
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Biobix, Lab of Bioinformatics and Computational Genomics, Department of Mathematical Modelling, Statistics and Bioinformatics, Ghent University, Ghent, Belgium
| | - Ine Koeken
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Behrouz Hassannia
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tom Vanden Berghe
- Cell Death Signaling lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
43
|
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
|
44
|
Wang Y, Liao B, Shan X, Ye H, Wen Y, Guo H, Xiao F, Zhu H. Revealing rutaecarpine's promise: A pathway to parkinson's disease relief through PPAR modulation. Int Immunopharmacol 2025; 147:114076. [PMID: 39809102 DOI: 10.1016/j.intimp.2025.114076] [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: 10/23/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
The pathological mechanisms of Parkinson's disease (PD) is complex, and no definitive cure currently exists. This study identified Rutaecarpine (Rut), an alkaloid extracted from natural plants, as a potential therapeutic agent for PD. To elucidate its mechanisms of action and specific effects in PD, network pharmacology, molecular docking, and experimental validation methods were employed. Our findings demonstrated the efficacy of Rut in ameliorating PD symptoms. Network pharmacology analysis indicated that Rut exerts its therapeutic effects through the PPAR signaling pathway and the lipid pathway. Molecular docking results revealed that Rut forms stable protein-ligand complexes with PPARα and PPARγ. Animal experiments showed that Rut improved motor function in PD mice, protected dopaminergic neurons, ameliorated lipid metabolism disorders, and reduced neuroinflammation. This study identified the critical molecular mechanisms and therapeutic targets of Rut in the treatment of PD, providing a theoretical foundation for future investigations into the pharmacodynamics of Rut as a potential anti-PD agent.
Collapse
Affiliation(s)
- Yeying Wang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; The Second Clinical Medical College of Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Bin Liao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Xuesong Shan
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Haonan Ye
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Yuqi Wen
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Feng Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Hong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| |
Collapse
|
45
|
Lee J, Roh JL. Lipid metabolism in ferroptosis: Unraveling key mechanisms and therapeutic potential in cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189258. [PMID: 39746458 DOI: 10.1016/j.bbcan.2024.189258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/29/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Ferroptosis, a form of iron-dependent cell death driven by lipid peroxidation, has emerged as a critical area of research for cancer therapy. This review delves into the intricate relationship between lipid metabolism and ferroptosis, emphasizing the impact of lipidome remodeling on cancer cell susceptibility. We explore key mechanisms, such as the role of polyunsaturated fatty acids and phosphatidylethanolamines in ferroptosis induction, alongside the protective effects of monounsaturated fatty acids and their regulatory enzymes. We also discuss the influence of dietary fatty acids, lipid droplets, and the epithelial-to-mesenchymal transition on ferroptosis and cancer resistance. By integrating current findings on enzymatic regulation, lipid peroxidation pathways, and metabolic adaptations, this review highlights potential therapeutic strategies targeting lipid metabolism to enhance ferroptosis-based cancer treatments. Our goal is to provide a comprehensive overview that underscores the significance of lipid metabolic pathways in ferroptosis and their implications for developing novel cancer therapies.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea; Department of Biomedical Science, General Graduate School, CHA University, Pocheon, Republic of Korea.
| |
Collapse
|
46
|
Liu H, Zhang C, Li S, Wang S, Xiao L, Chen J, Xia C, Dai X. Overexpression Bcl-2 alleviated ferroptosis induced by molybdenum and cadmium co-exposure through inhibiting mitochondrial ROS in duck kidneys. Int J Biol Macromol 2025; 291:139118. [PMID: 39719230 DOI: 10.1016/j.ijbiomac.2024.139118] [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: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/21/2024] [Indexed: 12/26/2024]
Abstract
Excessive molybdenum (Mo) and cadmium (Cd) are environmental pollutants with serious nephrotoxicity. B-cell lymphoma 2 (Bcl-2) plays a critical role in modulating mitochondrial ROS (Mito-ROS). Ferroptosis is a form of cell death dependent on lipid peroxidation. However, the impacts of Mo and Cd co-exposure on ferroptosis in duck kidneys and the function of Bcl-2 in the process are still unclear. Ducks and duck primary renal tubular epithelial cells exposed to different doses of Mo and/or Cd were used as the research target. Our work suggested that Mo and/or Cd significantly decreased Bcl-2 protein level and induced ferroptosis with the increase of ferrous ion, lipid peroxidation, TF protein level and the decrease of GPX4, FT protein levels. The Bcl-2 inhibitor HA14-1 exacerbated the changes of these indexes, but Bcl-2 overexpression had the opposite effect. Mito-ROS inhibitor ROS-IN-1 alleviated ferroptosis induced by Mo and Cd. Besides, Bcl-2 was involved in mitochondrial dysfunction induced by Mo and Cd, accompanied by disturbing Mito-ROS, ATP level, mitochondrial complex IV activity, Bcl-2 and COX-2 co-localization, lipid peroxidation, mitochondrial membrane potential (MMP) and mitochondrial structure. These findings substantiated that overexpression Bcl-2 alleviated ferroptosis co-induced by Mo and Cd through reducing Mito-ROS level in duck kidneys.
Collapse
Affiliation(s)
- Hang Liu
- College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Caiying Zhang
- College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Shanxin Li
- College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Sunan Wang
- College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Li Xiao
- College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Jirong Chen
- College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Chenjie Xia
- College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Xueyan Dai
- College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China.
| |
Collapse
|
47
|
Jang N, Kim IK, Jung D, Chung Y, Kang YP. Regulation of Ferroptosis in Cancer and Immune Cells. Immune Netw 2025; 25:e6. [PMID: 40078787 PMCID: PMC11896659 DOI: 10.4110/in.2025.25.e6] [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: 12/06/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
Ferroptosis, an iron-dependent form of regulated cell death, is driven by lipid peroxidation and shaped by metabolic and antioxidant pathways. In immune cells, ferroptosis susceptibility varies by cell types, lipid composition, and metabolic demands, influencing immune responses in cancer, infections, and autoimmune diseases. Therapeutically, targeting ferroptosis holds promise in cancer immunotherapy by enhancing antitumor immunity or inhibiting immunosuppressive cells. This review highlights the metabolic pathways underlying ferroptosis, its regulation in immune cells, its dual role in tumor progression and antitumor immunity, and its context-dependent therapeutic implications for optimizing cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Yeonseok Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
48
|
Lv S, Luo C. Ferroptosis in schizophrenia: Mechanisms and therapeutic potentials (Review). Mol Med Rep 2025; 31:37. [PMID: 39611491 PMCID: PMC11613623 DOI: 10.3892/mmr.2024.13402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Schizophrenia, a complex psychiatric disorder, presents with multifaceted symptoms and important challenges in treatment, primarily due to its pathophysiological complexity, which involves oxidative stress and aberrant iron metabolism. Recent insights into ferroptosis, a unique form of iron‑dependent cell death characterized by lipid peroxidation and antioxidant system failures, open new avenues for understanding the neurobiological foundation of schizophrenia. The present review explores the interplay between ferroptosis and schizophrenia, emphasizing the potential contributions of disrupted iron homeostasis and oxidative mechanisms to the pathology and progression of this disease. The emerging evidence linking ferroptosis with the oxidative stress observed in schizophrenia provides a compelling narrative for re‑evaluating current therapeutic strategies and exploring novel interventions targeting these molecular pathways, such as the glutathione peroxidase 4 pathway and the ferroptosis suppressor protein 1 pathway. By integrating recent advances in ferroptosis research, the current review highlights innovative therapeutic potentials, including N‑acetylcysteine, selenium, omega‑3 fatty acids and iron chelation therapy, which could address the limitations of existing treatments and improve clinical outcomes for individuals with schizophrenia.
Collapse
Affiliation(s)
- Shuang Lv
- Department of Psychiatry, Guangzhou Kangning Hospital (The Psychiatric Hospital of Guangzhou Civil Administration Bureau), Guangzhou, Guangdong 510430, P.R. China
| | - Chunxia Luo
- Department of Psychiatry, Guangzhou Kangning Hospital (The Psychiatric Hospital of Guangzhou Civil Administration Bureau), Guangzhou, Guangdong 510430, P.R. China
| |
Collapse
|
49
|
Zeng W, Zhang R, Huang P, Chen M, Chen H, Zeng X, Liu J, Zhang J, Huang D, Lao L. Ferroptotic Neutrophils Induce Immunosuppression and Chemoresistance in Breast Cancer. Cancer Res 2025; 85:477-496. [PMID: 39531510 PMCID: PMC11786957 DOI: 10.1158/0008-5472.can-24-1941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/28/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Inducing ferroptosis in tumor cells is emerging as a strategy for treating malignancies that are refractory to traditional treatment modalities. However, the consequences of ferroptosis of immune cells in the tumor microenvironment need to be better understood in order to realize the potential of this approach. In this study, we discovered that neutrophils in chemoresistant breast cancer are highly sensitive to ferroptosis. Reduction of the acyltransferase MOAT1 in chemoresistance-associated neutrophils induced phospholipid reprogramming, switching the preference from monounsaturated fatty acids to polyunsaturated fatty acids, which increased their susceptibility to ferroptosis. Ferroptotic neutrophils secreted PGE2, IDO, and oxidized lipids that suppressed the proliferation and cytotoxicity of antitumor CD8+ T cells. Furthermore, neutrophil ferroptosis was closely related to a distinct subset of IL1β+CXCL3+CD4+ (Fer-CD4) T lymphocytes, which were enriched in chemoresistant tumors. Fer-CD4 T cells orchestrated neutrophil ferroptosis by modulating MOAT1 expression via IL1β/IL1R1/NF-κB signaling. Moreover, Fer-CD4 T cells secreted CXCL3, IL8, and S100A9 to replenish the neutrophil pool in the tumor microenvironment. Ferroptotic neutrophils in turn fostered Fer-CD4 T-cell differentiation. In spontaneous tumorigenesis mouse models, targeting IL1β+ CD4+ T cells or IL1R1+ neutrophils broke the cross-talk, restraining neutrophil ferroptosis, enhancing antitumor immunity, and overcoming chemoresistance. Overall, these findings uncover the role of neutrophil ferroptosis in shaping the immune landscape and propose appealing targets for restoring immunosurveillance and chemosensitivity in breast cancer. Significance: In chemoresistant breast cancer, IL1β+CXCL3+CD4+ T cells mediate neutrophil ferroptosis that suppresses antitumor immunity, indicating that interfering with this intercellular cross-talk could be an attractive strategy to reverse chemoresistance.
Collapse
Affiliation(s)
- Wenfeng Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ruihua Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Penghan Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Minxia Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Houying Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xin Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiang Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiahui Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Di Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Liyan Lao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
50
|
Huang X, Lin K, Chen W, Zhang D, Khan M, Ye X, Wang B, Chen C, Tian Y, Yuan Y, Lin J. Modulation of the local angiotensin II: Suppression of ferroptosis and radiosensitivity in nasopharyngeal carcinoma via the HIF-1α-HILPDA axis. Radiother Oncol 2025; 203:110686. [PMID: 39709027 DOI: 10.1016/j.radonc.2024.110686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 11/06/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
PURPOSE Radiotherapy presents a curative approach for nasopharyngeal carcinoma (NPC); however, the cellular radiosensitivity heterogeneity limits its efficacy. Thus, investigating the specific mechanisms of radioresistance in NPC is crucial for identifying and employing effective radiosensitizing agents to enhance treatment success. METHODS AND MATERIALS Radioresistant NPC cell lines HONE1-RR and SUNE1-RR were established. Quantitative reverse transcription-PCR (qRT-PCR), western blot, and enzyme-linked immuno sorbent assay (ELISA) were employed to detect the activation of the angiotensinogen (AGT) and local angiotensin II (Ang II). Transmission electron microscopy, ferrous ion detection, and lipid oxidation levels were utilized to detect radiation-induced ferroptosis in NPC. Bioinformatics analysis, along with qRT-PCR, western blotting, co-immunoprecipitation, and dual-luciferase assays were employed to explore downstream mechanisms. Colony formation assay, Cell Counting Kit-8 (CCK-8) assay, and a nude mouse xenograft model were utilized to assess NPC radiosensitivity. The expression of AGT, hypoxia-inducible factor-1 alpha (HIF-1α), hypoxia-inducible lipid droplet-associated protein (HILPDA), and glutathione peroxidase 4 (GPX4) in NPC tissues was detected through immunohistochemistry. RESULTS Activation of local Ang II was revealed to play a critical role in driving radioresistance in NPC cells modulating ferroptosis. This local Ang II established a positive feedback loop with HIF-1α through two parallel pathways; Ang II stabilizes HIF-1α by activating the MAPK pathway, and AGT directly binds HIF-1α to prevent its degradation. This AGT-HIF-1α loop regulated NPC cell ferroptosis via transcriptional regulation of HILPDA expression. Moreover, the co-administration of Ang II receptor antagonist (ARB) and ferroptosis inducers markedly increased NPC radiosensitivity.Additionally, the expression of AGT, HIF-1α, and HILPDA was closely correlated with the intensity of ferroptosis, radiosensitivity, and prognosis in NPC. CONCLUSIONS Our findings suggest that the AGT-HIF-1α-HILPDA pathway promotes radioresistance in NPC by enhancing lipid droplet accumulation, thereby suppressing ferroptosis. Targeting local Ang II alongside ferroptosis induction offers a promising strategy to improve radiosensitivity in NPC.
Collapse
Affiliation(s)
- Xiuting Huang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Kehai Lin
- Department of Oncology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000, China
| | - Weirui Chen
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Donghui Zhang
- Department of Pathology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Muhammad Khan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Xiaoxin Ye
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Baiyao Wang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Chengcong Chen
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China
| | - Yunhong Tian
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China.
| | - Yawei Yuan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China.
| | - Jie Lin
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510000, China.
| |
Collapse
|