1
|
Kato K, Yasui H, Sato-Akaba H, Emoto MC, Fujii HG, Kmiec MM, Kuppusamy P, Nagane M, Yamashita T, Inanami O. Non-invasive electron paramagnetic resonance imaging detects tumor redox imbalance induced by ferroptosis. Redox Rep 2025; 30:2454887. [PMID: 39836064 PMCID: PMC11753017 DOI: 10.1080/13510002.2025.2454887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025] Open
Abstract
Targeting ferroptosis, cell death caused by the iron-dependent accumulation of lipid peroxides, and disruption of the redox balance are promising strategies in cancer therapy owing to the physiological characteristics of cancer cells. However, the detection of ferroptosis using in vivo imaging remains challenging. We previously reported that redox maps showing the reduction power per unit time of implanted tumor tissues via non-invasive redox imaging using a novel, compact, and portable electron paramagnetic resonance imaging (EPRI) device could be compared with tumor tissue sections. This study aimed to apply the EPRI technique to the in vivo detection of ferroptosis. Notably, redox maps reflecting changes in the redox status of tumors induced by the ferroptosis-inducing agent imidazole ketone erastin (IKE) were compared with the immunohistochemical images of 4-hydroxynonenal (4-HNE) in tumor tissue sections. Our comparison revealed a negative correlation between the reducing power of tumor tissue and the number of 4-HNE-positive cells. Furthermore, the control and IKE-treated groups exhibited significantly different distributions on the correlation map. Therefore, redox imaging using EPRI may contribute to the non-invasive detection of ferroptosis in vivo.
Collapse
Affiliation(s)
- Kazuhiro Kato
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Hideo Sato-Akaba
- Department of Electrical and Electronic Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Hamamatsu, Japan
| | - Miho C. Emoto
- Department of Clinical Laboratory Science, School of Medical Technology, Health Sciences University of Hokkaido, Sapporo, Japan
| | - Hirotada G. Fujii
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari, Japan
| | - Maciej M. Kmiec
- Departments of Radiology and Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Periannan Kuppusamy
- Departments of Radiology and Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Masaki Nagane
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Tadashi Yamashita
- Laboratory of Biochemistry, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Department of Applied Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
2
|
Zheng Q, Wang D, Lin R, Xu W. Pyroptosis, ferroptosis, and autophagy in spinal cord injury: regulatory mechanisms and therapeutic targets. Neural Regen Res 2025; 20:2787-2806. [PMID: 39101602 PMCID: PMC11826477 DOI: 10.4103/nrr.nrr-d-24-00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/24/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Regulated cell death is a form of cell death that is actively controlled by biomolecules. Several studies have shown that regulated cell death plays a key role after spinal cord injury. Pyroptosis and ferroptosis are newly discovered types of regulated cell deaths that have been shown to exacerbate inflammation and lead to cell death in damaged spinal cords. Autophagy, a complex form of cell death that is interconnected with various regulated cell death mechanisms, has garnered significant attention in the study of spinal cord injury. This injury triggers not only cell death but also cellular survival responses. Multiple signaling pathways play pivotal roles in influencing the processes of both deterioration and repair in spinal cord injury by regulating pyroptosis, ferroptosis, and autophagy. Therefore, this review aims to comprehensively examine the mechanisms underlying regulated cell deaths, the signaling pathways that modulate these mechanisms, and the potential therapeutic targets for spinal cord injury. Our analysis suggests that targeting the common regulatory signaling pathways of different regulated cell deaths could be a promising strategy to promote cell survival and enhance the repair of spinal cord injury. Moreover, a holistic approach that incorporates multiple regulated cell deaths and their regulatory pathways presents a promising multi-target therapeutic strategy for the management of spinal cord injury.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Weihong Xu
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
3
|
Yu H, Zhou C, Yang S, Yu J, Zhang X, Liang Z, Tan S, Song Y, Wang W, Sun Y, Zan R, Qiu H, Shen L, Zhang X. Mitigation of arteriosclerosis through transcriptional regulation of ferroptosis and lipid metabolism by magnesium. Biomaterials 2025; 319:123135. [PMID: 39985976 DOI: 10.1016/j.biomaterials.2025.123135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/24/2025]
Abstract
Metallic cardiovascular stents are crucial for preventing atherosclerosis-induced infarction by offering mechanical support. However, the effects of metal ions released from these stents on atherosclerosis remain ambiguous. This study evaluates the potential impact posed by the degradation products of magnesium-based stents, with a focus on ferroptosis, a key mechanism driving atherosclerosis. Remarkably, our results demonstrate that Mg effectively inhibits ferroptosis in human umbilical vein endothelial cells and in murine, rat and rabbit models. Our studies reveal that magnesium ions impede the dephosphorylation of ERK proteins, thereby enhancing the expression of SLC7A11 and GCL proteins via activation of the MAPK pathway mechanistically. Additionally, magnesium ions downregulate ACSL4 protein expression, leading to decreased levels of acyl-CoA and ether-phospholipids. Eventually, multiple animal experiments indicate that biodegradable Mg stents can inhibit ferroptosis and decelerate the progression of arteriosclerosis, highlighting the therapeutic potential of Mg stents in treating arteriosclerosis.
Collapse
Affiliation(s)
- Han Yu
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Changyi Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200030, China
| | - Shi Yang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jinlong Yu
- Orthopaedic Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xiyue Zhang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhaojia Liang
- Stomatologic Hospital and College, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Shuang Tan
- Stomatologic Hospital and College, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Yang Song
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wenhui Wang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yu Sun
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rui Zan
- Yiwu Research Institute of Fudan University, Fudan University, Yiwu, Zhejiang, 322000, China.
| | - Hua Qiu
- Stomatologic Hospital and College, Key Laboratory of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, Anhui, 230032, China.
| | - Li Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200030, China.
| | - Xiaonong Zhang
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
4
|
Ma LF, Luo SR, Liu QQ, Tang RS, Chen NY, Luo D, Guo L, Li J, Wu R, Zhan ZJ. Neuroprotective lindenane sesquiterpenoids from the roots of Lindera aggregata (Sims) Kosterm. PHYTOCHEMISTRY 2025; 235:114452. [PMID: 39986409 DOI: 10.1016/j.phytochem.2025.114452] [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: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Eleven previously undescribed lindenane sesquiterpenoids, lindaggrols A-K (1-11), were isolated from the roots of Lindera aggregata (Sims) Kosterm, together with five known ones. Their structures were elucidated by HR-ESI-MS, NMR, and single-crystal X-ray diffraction analyses. Lindaggrol A (1) is an undescribed rearranged dinor-lindenane with an unprecedented 3/5/5 tricyclic scaffold. All isolated compounds were assayed for their neuroprotective effects against erastin-induced ferroptosis in HT-22 cells. Among them, 3, 12 and 14 exhibited significant neuroprotective activities with EC50 values ranging from 1.4 to 8.7 μM.
Collapse
Affiliation(s)
- Lie-Feng Ma
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR China
| | - Shan-Rong Luo
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR China
| | - Qian-Qing Liu
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR China
| | - Rui-Si Tang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Ning-Yu Chen
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR China
| | - Di Luo
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR China
| | - Lu Guo
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR China
| | - Jia Li
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Rui Wu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China.
| | - Zha-Jun Zhan
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR China.
| |
Collapse
|
5
|
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
|
6
|
Rani K, Chand Sahu R, Chaudhuri A, Kumar DN, Arora S, Kumar D, Agrawal AK. Exploring combinations of dihydroartemisinin for cancer therapy: A comprehensive review. Biochem Biophys Res Commun 2025; 765:151854. [PMID: 40262468 DOI: 10.1016/j.bbrc.2025.151854] [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: 01/07/2025] [Revised: 03/22/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025]
Abstract
Cancer remains a significant threat to human health due to its multifaceted causes and complex pathogenesis. While advancements in research have improved outcomes for many cancer patients, treatments for specific tumor types still face limitations. Dihydroartemisinin (DHA), an active metabolite of artemisinin and its derivatives, has proven to be an effective anti-malarial agent. Recently, its anticancer potential has garnered increasing interest as it acts through multiple molecular pathways, including anti-proliferation, induction of apoptosis, autophagy and endoplasmic reticulum (ER) stress, anti-metastasis, inhibition of angiogenesis, and modulation of immune function. This review aims to thoroughly explain and summarize the mechanisms of DHA against cancer and the latest progress in this field. Due to the insufficiency of monotherapy in effectively treating cancer, the use of chemotherapy in combination with alternative therapies has witnessed a notable increase in popularity. DHA has shown synergistic anti-tumor efficacy with a range of therapeutic drugs, but its co-delivery with chemotherapeutics has been limited by low solubility and bioavailability. Nanotechnology-assisted co-delivery of anti-tumor agents, utilizing advanced stimulus-triggered drug release systems in tumor cells, offers the potential to enhance selective delivery and increase antitumor efficacy. Additionally, this article provides suggestions for further research on the anticancer effects of DHA.
Collapse
Affiliation(s)
- Komal Rani
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, India
| | - Rohan Chand Sahu
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, India
| | - Aiswarya Chaudhuri
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, India
| | - Dulla Naveen Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, India
| | - Sanchit Arora
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, India
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, India
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, UP, India.
| |
Collapse
|
7
|
Gong L, Wu L, Zhao S, Xiao S, Chu X, Zhang Y, Li F, Li S, Yang H, Jiang P. Epigenetic regulation of ferroptosis in gastrointestinal cancers (Review). Int J Mol Med 2025; 55:93. [PMID: 40242977 PMCID: PMC12045471 DOI: 10.3892/ijmm.2025.5534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Ferroptosis is a type of iron‑dependent cell death characterized by excessive lipid peroxidation and may serve as a potential therapeutic target in cancer treatment. While the mechanisms governing ferroptosis continue to be explored and elucidated, an increasing body of research highlights the significant impact of epigenetic modifications on the sensitivity of cancer cells to ferroptosis. Epigenetic processes, such as DNA methylation, histone modifications and non‑coding RNAs, have been identified as key regulators that modulate the expression of ferroptosis‑related genes. These alterations can either enhance or inhibit the sensitivity of gastrointestinal cancer (GIC) cells to ferroptosis, thereby affecting the fate of GICs. Drugs that target epigenetic markers for advanced‑stage cancer have shown promising results in enhancing ferroptosis and inhibiting tumor growth. This review explores the intricate relationship between epigenetic regulation and ferroptosis in GICs. Additionally, the potential of leveraging epigenetic modifications to trigger ferroptosis in GICs is investigated. This review highlights the importance of further research to elucidate the specific mechanisms underlying epigenetic control of ferroptosis and to advance the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Linqiang Gong
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Linlin Wu
- Oncology Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shiyuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| | - Shuai Xiao
- Department of Intensive Care Medicine, Tengzhou Central People's Hospital, Jining Medical University, Tengzhou, Shandong 277500, P.R. China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
| | - Yazhou Zhang
- Department of Foot and Ankle Surgery, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Fengfeng Li
- Neurosurgery Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shuhui Li
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Hui Yang
- Department of Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| |
Collapse
|
8
|
Wu Y, Zhu BT. Role of protein disulfide isomerase in mediating sulfasalazine-induced ferroptosis in HT22 cells: The PDI-NOS-NO-ROS/lipid-ROS cascade. Arch Biochem Biophys 2025; 768:110366. [PMID: 40023379 DOI: 10.1016/j.abb.2025.110366] [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: 11/03/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Ferroptosis is a form of regulated cell death resulting from excessive lipid peroxidation. Sulfasalazine (SAS), an anti-inflammatory drug, can induce ferroptosis through inhibiting the system Xc- and triggering glutathione depletion. SAS has attracted considerable interest in recent years because of its potential for repurposing as an anticancer agent. Our recent studies have shown that protein disulfide isomerase (PDI) is an upstream mediator of chemically-induced ferroptosis through catalyzing the dimerization of nitric oxide synthase (NOS) and NO accumulation in cultured HT22 hippocampal neuronal cells. The present study aims to investigate SAS-induced ferroptotic cell death in HT22 cells with a focus on determining the role of PDI in mediating SAS-induced ferroptosis. We find that SAS induces ferroptotic cell death in HT22 cells, which is accompanied by a time-dependent sequential increase in the accumulation of cellular NO, ROS and lipid-ROS. We find that treatment of HT22 cells with SAS activates PDI-mediated iNOS activation (dimerization) and NO accumulation. In addition, SAS also strongly upregulates iNOS protein levels in HT22 cells. PDI knockdown or pharmacological inhibition of PDI's activity each suppresses SAS-induced iNOS dimerization, which is associated with abrogation of SAS-induced accumulation of NO, ROS and lipid-ROS, and a strong protection against ferroptotic cell death. On the other hand, PDI activation through the use of a TrxR1 inhibitor can strongly sensitize cells to SAS-induced ferroptosis. Together, these experimental observations demonstrate a crucial role of PDI in SAS-induced ferroptosis in a cell culture model through the activation of the PDI → NOS → NO → ROS/lipid-ROS pathway. Insights gained from this study also provide effective strategies to selectively sensitizing human cancer cells to SAS-induced ferroptosis, such as through the use of NO-releasing agents or TrxR1 inhibitors.
Collapse
Affiliation(s)
- Yufei Wu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China; Shenzhen Bay Laboratory, Shenzhen, 518172, China.
| |
Collapse
|
9
|
Yang H, Yuan D, Zhou Z, Zhao H. Nitrate enrichment exacerbates microbiome and metabolism disturbances of the coral holobiont under heat stress. MARINE ENVIRONMENTAL RESEARCH 2025; 208:107098. [PMID: 40139064 DOI: 10.1016/j.marenvres.2025.107098] [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: 01/19/2025] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Coral reef ecosystems are facing severe deterioration due to escalating global temperatures and human-induced activities. Combined nitrate and heat stress can exacerbate coral bleaching, however, the underlying mechanism is still unclear. In the present study, we assessed the bleaching status of Acropora hyacinthus, a reef-building coral species, under high temperature and nitrate stress conditions using chemostat cultivation. We observed nitrate enrichment (9 μM) induced a significant reduction in photosystem efficiency (Fv/Fm) of Symbiodiniaceae and an increased thermal bleaching of corals under high temperature (30 °C). Nitrate exposure promoted the proliferation of Enterobacteriaceae and Vibrionaceae, which are bacterial families, potentially augmenting the coral's susceptibility to disease while exerting negligible effects on the fungal community. Alterations were observed in the metabolic pathways of both the coral hosts and Symbiodiniaceae, including down-regulated folate biosynthesis and inflammatory mediator regulation of TRP channels. Our findings indicate that nitrate enrichment under heat stress disrupts the metabolism of coral holobionts through altering bacterial communities, ultimately leading to increased coral bleaching.
Collapse
Affiliation(s)
- Huidan Yang
- Hainan International Joint Research Center for Coral Reef Ecology, Hainan University, Haikou, 570228, China; Center for Eco-Environment Restoration of Hainan Province, School of Ecology, Hainan University, Haikou, 570228, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, China
| | - Dongdan Yuan
- Hainan International Joint Research Center for Coral Reef Ecology, Hainan University, Haikou, 570228, China; Center for Eco-Environment Restoration of Hainan Province, School of Ecology, Hainan University, Haikou, 570228, China
| | - Zhuojing Zhou
- Hainan International Joint Research Center for Coral Reef Ecology, Hainan University, Haikou, 570228, China; Center for Eco-Environment Restoration of Hainan Province, School of Ecology, Hainan University, Haikou, 570228, China
| | - Hongwei Zhao
- Hainan International Joint Research Center for Coral Reef Ecology, Hainan University, Haikou, 570228, China; Center for Eco-Environment Restoration of Hainan Province, School of Ecology, Hainan University, Haikou, 570228, China.
| |
Collapse
|
10
|
Chen J, Fu Y, Chen S, Weng S, He J, Dong C. Scale drop disease virus (SDDV) triggers ferroptosis both in mandarin fish (Siniperca chuatsi) and MFF-1 cells to facilitate virus infection via linking to transferrin receptor 1 (TfR1). FISH & SHELLFISH IMMUNOLOGY 2025; 163:110416. [PMID: 40373888 DOI: 10.1016/j.fsi.2025.110416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 05/08/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
Scale drop disease virus (SDDV) is a distinct member in genus Megalocytivirus of family Iridoviridae, garnering increasing attention due to its significant threat to teleost. Ferroptosis is a new type of cell death discovered recently and involved in various viral infections. Knowledges on SDDV induced ferroptosis remains unclear. Here, we demonstrated that SDDV infection triggers ferroptosis, as evidenced by hallmark features such as iron overload, massive lipid peroxides accumulation, glutathione depletion and glutathione peroxidase 4 (gpx4) downregulation. SDDV-infected MFF-1 cells exhibited increased reactive oxygen species production and mitochondrial shrinkage. Treatment with Ferrostatin-1, a potent ferroptosis inhibitor, significantly attenuated SDDV replication in MFF-1 cells and could improve the survival of mandarin fish upon SDDV challenge. Treatment with an iron chelator mitigated ferroptosis and reduced the mortality of mandarin fish following SDDV infection, suggesting that SDDV-induced ferroptosis is iron-dependent. Finally, we demonstrated that SDDV infection could upregulate the expression of transferrin receptor protein 1 (TfR1), a critical iron transporter, to disrupt cellular iron homeostasis, induce ferroptosis, and then facilitate viral infection. Collectively, our findings provide compelling evidence that SDDV infection induces ferroptosis by targeting TfR1 to facilitate virus infection. Inhibiting ferroptosis maybe represent a promising anti-viral strategy for combating SDDV infection in aquaculture.
Collapse
Affiliation(s)
- Jiaming Chen
- State Key Laboratory of Biocontrol/School of Life Sciences of Sun Yat-sen University, Guangzhou, 510275, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Yuting Fu
- State Key Laboratory of Biocontrol/School of Life Sciences of Sun Yat-sen University, Guangzhou, 510275, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Shuyi Chen
- State Key Laboratory of Biocontrol/School of Life Sciences of Sun Yat-sen University, Guangzhou, 510275, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol/School of Life Sciences of Sun Yat-sen University, Guangzhou, 510275, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Jianguo He
- State Key Laboratory of Biocontrol/School of Life Sciences of Sun Yat-sen University, Guangzhou, 510275, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Chuanfu Dong
- State Key Laboratory of Biocontrol/School of Life Sciences of Sun Yat-sen University, Guangzhou, 510275, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, 510275, PR China.
| |
Collapse
|
11
|
Jwa NS, Hwang BK. Ferroptosis in plant immunity. PLANT COMMUNICATIONS 2025; 6:101299. [PMID: 40057824 DOI: 10.1016/j.xplc.2025.101299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/15/2025] [Accepted: 03/05/2025] [Indexed: 04/13/2025]
Abstract
Plant cell death is mediated by calcium, iron, and reactive oxygen species (ROS) signaling in plant immunity. The reconstruction of a nucleotide-binding leucine-rich-repeat receptor (NLR) supramolecular structure, called the resistosome, is intimately involved in the hypersensitive response (HR), a type of cell death involved in effector-triggered immunity (ETI). Iron is a crucial redox catalyst in various cellular reactions. Ferroptosis is a regulated, non-apoptotic form of iron- and ROS-dependent cell death in plants. Pathogen infections trigger iron accumulation and ROS bursts in plant cells, leading to lipid peroxidation via the Fenton reaction and subsequent ferroptosis in plant cells similar to that in mammalian cells. The small-molecule inducer erastin triggers iron-dependent lipid ROS accumulation and glutathione depletion, leading to HR cell death in plant immunity. Calcium (Ca2+) is another major mediator of plant immunity. Cytoplasmic Ca2+ influx through calcium-permeable channels, the resistosomes, mediates iron- and ROS-dependent ferroptotic cell death under reduced glutathione reductase (GR) expression levels in the ETI response. Acibenzolar-S-methyl (ASM), a plant defense activator, enhances Ca2+ influx, ROS and iron accumulation, and lipid peroxidation to trigger ferroptotic cell death. These breakthroughs suggest a potential role for Ca2+ signaling in ferroptosis and its coordination with iron and ROS signaling in plant immunity. In this review, we highlight the essential roles of calcium, iron, and ROS signaling in ferroptosis during plant immunity and discuss advances in the understanding of how Ca2+-mediated ferroptotic cell death orchestrates effective plant immune responses against invading pathogens.
Collapse
Affiliation(s)
- Nam-Soo Jwa
- Division of Integrative Bioscience and Biotechnology, College of Life Sciences, Sejong University, Seoul 05006, Korea.
| | - Byung Kook Hwang
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea.
| |
Collapse
|
12
|
Liu Z, Zhao Z, Xiao Z, Li M, Wang X, Huang Y, Li Y. Extracellular vesicles derived from bone marrow mesenchymal stem cells regulate SREBF2/HMGB1 axis by transporting miR-378a-3p to inhibit ferroptosis in intestinal ischemia-reperfusion injury. Cell Death Discov 2025; 11:223. [PMID: 40335466 PMCID: PMC12058992 DOI: 10.1038/s41420-025-02509-6] [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: 07/10/2024] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025] Open
Abstract
Intestinal ischemia-reperfusion (II/R) injury represents a life-threatening and complex pathophysiological process that remains challenging to treat clinically, and emerging evidence suggests that ferroptosis plays an essential role in its pathogenesis. This study aimed to investigate whether extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSC-EVs) can mitigate II/R-induced ferroptosis in a murine model. Using a bioinformatics database, we initially identified genes with abnormal expression patterns in II/R injury. Then, we confirmed the association between II/R injury, ferroptosis, and the HMGB1/SREBF2 axis through in vivo and in vitro experiments. To determine the role of HMGB1 in hypoxia/reoxygenation (H/R)-induced ferroptosis in Caco-2 cells, we transfected cells with either sh-HMGB1 or control sh-NC constructs and developed an H/R model in vitro. Subsequently, we examined factors regulating HMGB1-mediated ferroptosis in Caco-2 cells and assessed the effect of BMSC-EVs on this process. To further explore the mechanism underlying the protective effects of BMSC-EVs in II/R injury, we screened for miRNAs with reduced expression during II/R and verified their involvement. Among these, miR-378a-3p was identified as a candidate for regulating ferroptosis. To confirm its functional role, we treated II/R mice with BMSC-EVs overexpressing miR-378a-3p and assessed the outcomes. Our findings revealed that HMGB1, which is a key regulatory factor of ferroptosis, was significantly upregulated during II/R injury, and its knockdown alleviated H/R-induced ferroptosis in Caco-2 cells. We also found that SREBF2 directly regulates HMGB1 expression to promote H/R-induced ferroptosis in vitro. Importantly, BMSC-EVs alleviated II/R injury by suppressing ferroptosis in Caco-2 cells, and mechanistically, miR-378a-3p, a miRNA derived from BMSC-EVs, inhibited II/R-induced ferroptosis by modulating the SREBF2/HMGB1 axis. In conclusion, BMSC-EVs may exert protective effects against II/R injury by delivering miR-378a-3p, which regulates the SREBF2/HMGB1 axis to suppress ferroptosis, providing important insights into the pathological mechanisms underlying II/R injury and potential therapeutic strategies for its management.
Collapse
Affiliation(s)
- Zan Liu
- Department of Pediatric Surgery, Clinical Research Center for Pediatric Solid Tumors in Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan children's hospital), Changsha, PR China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, College of Pharmacy, Changsha Medical University, Changsha, PR China
| | - Zitong Zhao
- Center of Reproductive Medicine, Changsha Hospital for Maternal and Child Health Care of Hunan Normal University, Changsha, PR China
| | - Zhenghui Xiao
- Emergency center of Hunan Children's Hospital, Changsha, Hunan, PR China
| | - Ming Li
- Department of Pediatric Surgery, Clinical Research Center for Pediatric Solid Tumors in Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan children's hospital), Changsha, PR China
| | - Xiyang Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Yan Huang
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, PR China.
- Hunan Provincial Key Laboratory of Neurorestoration, Changsha, Hunan, PR China.
| | - Yong Li
- Department of Pediatric Surgery, Clinical Research Center for Pediatric Solid Tumors in Hunan Province, Hunan Provincial Key Laboratory of Pediatric Orthopedics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan children's hospital), Changsha, PR China.
| |
Collapse
|
13
|
Hao X, Wang Y, Hou MJ, Yang YX, Liao L, Chen T, Wang P, Chen X, Zhu BT. Strong protection by bazedoxifene against chemically-induced ferroptotic neuronal death in vitro and in vivo. Cell Commun Signal 2025; 23:218. [PMID: 40336106 PMCID: PMC12060420 DOI: 10.1186/s12964-025-02209-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
Ferroptosis, a form of regulated cell death associated with glutathione depletion and excess lipid peroxidation, can be induced in cultured cells by chemicals (e.g., erastin and RSL3). It has been shown that protein disulfide isomerase (PDI) is a mediator of chemically-induced ferroptosis and also a crucial target for ferroptosis protection. The present study reports that bazedoxifene (BAZ), a selective estrogen receptor modulator, is an inhibitor of PDI and can strongly rescue neuronal cells from chemically-induced oxidative ferroptosis. We find that BAZ can directly bind to PDI and inhibit its catalytic activity. Computational modeling analysis reveals that BAZ forms a hydrogen bond with PDI's His256 residue. Inhibition of PDI by BAZ markedly reduces iNOS and nNOS dimerization (i.e., catalytic activation) and NO accumulation, and these effects of BAZ are associated with reductions in cellular ROS and lipid-ROS and protection against chemically-induced ferroptosis. In addition, the direct antioxidant activity of BAZ may also partially contribute to its protection against chemically-induced ferroptosis. In vivo animal experiments show that mice treated with BAZ are strongly protected against kainic acid-induced oxidative hippocampal neuronal injury and memory deficits. Together, these results reveal that BAZ is a potent inhibitor of PDI and can strongly protect against chemically-induced ferroptosis in hippocampal neurons both in vitro and in vivo. This work provides evidence for an estrogen receptor-independent, PDI-mediated novel mechanism of neuroprotection by BAZ.
Collapse
Affiliation(s)
- Xiangyu Hao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Yifan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Ming-Jie Hou
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Yong Xiao Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Lixi Liao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Tongxiang Chen
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Pan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Xiaojun Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
14
|
Yang X, Dong S, Xing C, Li C, Bo C, Meng X, Liu Z, Shao H, Li M, Jia Q. Ferroptosis is involved in the benzene-induced hematotoxicity via mitochondrial ROS-ferritinophagy pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 376:126379. [PMID: 40334736 DOI: 10.1016/j.envpol.2025.126379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/17/2025] [Accepted: 05/04/2025] [Indexed: 05/09/2025]
Abstract
Benzene, a common environmental contaminant that significantly impacts the hematopoietic system. Although benzene toxicity has been well documented, the exact molecular mechanisms involved remain unclear. This study aimed to explore the role of ferroptosis in benzene-induced hematotoxicity and uncover the underlying mechanisms. Rats exposed to benzene exhibited reduced peripheral blood cell counts, elevated serum iron concentrations, and increased expression of proteins associated with autophagy and ferroptosis within their bone marrow (BM) cells. In addition, inhibition of autophagy in benzene-exposed rats alleviated weight loss, peripheral blood cell abnormalities, iron dysregulation, and ferroptosis signaling activation. To further investigate the cellular mechanisms, we conducted in vitro experiments in which the benzene metabolite hydroquinone (HQ) was found to elicit ferroptosis and disrupt autophagy functionality in JHP cells. Meanwhile, the autophagy inhibitor 3-methyladenine (3-MA) alleviated these adverse effects. Additionally, HQ induced damage to mitochondria in JHP cells, as evidenced by a decline in mitochondrial membrane potential (MMP) and an increase in mitochondrial reactive oxygen species (mtROS). Collectively, our results demonstrate that mtROS-dependent autophagy participates in ferroptosis induced by benzene, providing a significant theoretical foundation for the pathogenesis and potential interventions underlying benzene-induced hematotoxicity.
Collapse
Affiliation(s)
- Xiaohan Yang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan, 250062, China
| | - Shuangyan Dong
- Jinan Changqing District Disease Prevention and Control Center, Ji'nan, 250399, China
| | - Caihong Xing
- Key Laboratory of Chemical Safety and Health, National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention (CDC), Beijing, 100050, China
| | - Chao Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan, 250062, China
| | - Cunxiang Bo
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan, 250062, China
| | - Xiangjing Meng
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan, 250062, China
| | - Zhidan Liu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan, 250062, China
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan, 250062, China
| | - Ming Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan, 250062, China.
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Science, Ji'nan, 250062, China; School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250000, China.
| |
Collapse
|
15
|
S Y, K L M, Harithpriya K, Zong C, Sahabudeen S, Ichihara G, Ramkumar KM. Disruptive multiple cell death pathways of bisphenol-A. Toxicol Mech Methods 2025; 35:430-443. [PMID: 39815394 DOI: 10.1080/15376516.2024.2449423] [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: 11/11/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025]
Abstract
Endocrine-disrupting chemicals (EDCs) significantly contribute to health issues by interfering with hormonal functions. Bisphenol A (BPA), a prominent EDC, is extensively utilized as a monomer and plasticizer in producing polycarbonate plastic and epoxy resins, making it one of the highest-demanded chemicals in commercial use. This is the major component used in plastic products, including bottles, containers, storage items, and food serving ware. Exposure of BPA happens through oral, respiratory, transdermal routes and eye contact. As an EDC, BPA disrupts hormonal binding, leading to various health problems, such as cancers, reproductive abnormalities, metabolic syndrome, immune dysfunction, neurological effects, cardiovascular problems, respiratory issues, and obesity. BPA mimics the hormone estrogen but exhibits a weak affinity for estrogen receptors. This weak binding affinity triggers multiple cell death pathways, including necroptosis, pyroptosis, apoptosis, ferroptosis, and autophagy, across different cell types. Numerous clinical, in-vitro, and in-vivo experiments have demonstrated that BPA exposure results in unfavorable health effects. This review highlights the mechanisms of cell death pathways initiated through BPA exposure and the associated negative health consequences. The extensive use of BPA and its frequent detection in environmental and biological models underscore the urgent need for further investigation into its effects and the development of safe alternatives. Addressing the health risks posed by BPA involves a comprehensive approach that includes reducing exposure and finding novel substitutes to lessen its detrimental impact on humans.
Collapse
Affiliation(s)
- Yukta S
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Milan K L
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Cai Zong
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Science, Tokyo University of Science, Tokyo, Japan
| | - S Sahabudeen
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Science, Tokyo University of Science, Tokyo, Japan
| | - K M Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, India
| |
Collapse
|
16
|
Lv Z, Wang J, Xu J, Chen X, Lu D, Huang J, Shen XC, Chen H. Protein-Interference-Free and Kidney-Targeting NIR Fluorophores for Accurate in Vivo Imaging of H 2S 2 during Kidney Ferroptosis. Adv Healthc Mater 2025; 14:e2500273. [PMID: 40125815 DOI: 10.1002/adhm.202500273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/04/2025] [Indexed: 03/25/2025]
Abstract
High-fidelity kidney function imaging is important for assessing the nephrotoxicity of drugs and diagnosing renal diseases. However, the current challenges in achieving accurate kidney imaging include unspecific signal enhancement due to albumin binding and relatively low distribution of imaging agents in kidneys. Here, for the first time, a side-chain engineering strategy that incorporates hydrophilic six-membered heterocycles into aza-hemicyanine for generating high-performance kidney imaging agents with protein-interference-free and kidney-targeting features is proposed. Based on these unique aza-hemicyanine dyes, the first kidney-targeting and albumin-insensitive H2S2 near-infrared (NIR) fluorescent probe NA-H2S2 is designed, which demonstrates effective kidney distribution following intravenous injection and is specifically activated by H2S2. The designed probe presents a highly rapid, selective and sensitive response to H2S2 with a detection limit as low as 24.21 nm. Additionally, it successfully achieves real-time in vivo NIR fluorescence imaging of H2S2 during erastin/cisplatin induced renal ferroptosis. Moreover, it also enables rapid detection of H2S2 through in vitro optical urinalysis, offering significant diagnostic value for renal ferroptosis. Overall, this study not only presents a practical kidney-targeting H2S2 fluorescent probe NA-H2S2 with increased imaging accuracy but also provides promising kidney-targeting and albumin-insensitive aza-hemicyanine dyes for further development of kidney disease-related probes.
Collapse
Affiliation(s)
- Zhangkang Lv
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Jing Wang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Jinyuan Xu
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Xiaoshan Chen
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Dongxue Lu
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Jingting Huang
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Xing-Can Shen
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| | - Hua Chen
- Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Guangxi Key Laboratory of Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, P. R. China
| |
Collapse
|
17
|
Le HT, Kim Y, Kim MJ, Hyun SH, Kim H, Chung SW, Joe Y, Chung HT, Shin DM, Back SH. Phosphorylation of eIF2α suppresses the impairment of GSH/NADPH homeostasis and mitigates the activation of cell death pathways, including ferroptosis, during ER stress. Mol Cells 2025; 48:100210. [PMID: 40089158 PMCID: PMC11999272 DOI: 10.1016/j.mocell.2025.100210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025] Open
Abstract
eIF2α Phosphorylation helps maintain cellular homeostasis and overcome endoplasmic reticulum (ER) stress through transcriptional and translational reprogramming. This study aims to elucidate the transcriptional regulation of glutathione (GSH) and nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) homeostasis through eIF2α phosphorylation and its impact on cell death during ER stress. eIF2α phosphorylation-deficient (A/A) cells exhibited decreased expression of multiple genes involved in GSH synthesis and NADPH production, leading to an exacerbated depletion of both cellular and mitochondrial GSH, as well as mitochondrial NADPH, during ER stress. Impaired GSH homeostasis resulted from deficient expression of ATF4 and/or its dependent factor, Nrf2, which are key transcription factors in the antioxidant response during ER stress. In contrast, the exacerbation of NADPH depletion may primarily be attributed to the dysregulated expression of mitochondrial serine-driven 1-carbon metabolism pathway genes, which are regulated by an unidentified eIF2α phosphorylation-dependent mechanism during ER stress. Moreover, the eIF2α phosphorylation-ATF4 axis was responsible for upregulation of ferroptosis-inhibiting genes and downregulation of ferroptosis-activating genes upon ER stress. Therefore, ER stress strongly induced ferroptosis of A/A cells, which was significantly inhibited by treatments with cell-permeable GSH and the ferroptosis inhibitor ferrostatin-1. ATF4 overexpression suppressed impairment of GSH homeostasis in A/A cells during ER stress by promoting expression of downstream target genes. Consequently, ATF4 overexpression mitigated ferroptosis as well as apoptosis of A/A cells during ER stress. Our findings underscore the importance of eIF2α phosphorylation in maintaining GSH/NADPH homeostasis and inhibiting ferroptosis through ATF4 and unidentified eIF2α phosphorylation-dependent target(s)-mediated transcriptional reprogramming during ER stress.
Collapse
Affiliation(s)
- Hien Thi Le
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Yonghwan Kim
- Department of Cell and Genetic Engineering, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Mi-Jeong Kim
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Seung Hwa Hyun
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Hyeeun Kim
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Su Wol Chung
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Yeonsoo Joe
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| | - Hun Taeg Chung
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Korea
| | - Dong-Myung Shin
- Department of Cell and Genetic Engineering, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea; Basic-Clinical Convergence Research Center, University of Ulsan, Ulsan 44610, Korea.
| |
Collapse
|
18
|
Lu Y, Ge Y, Tu F, Li X, Geng P, Zhang F, Wang Q. In Situ Investigation of Intercellular Communication in Ferroptosis Integrated Scanning Electrochemical Microscopy with Microfluidic Devices. ACS Sens 2025. [PMID: 40295188 DOI: 10.1021/acssensors.5c00176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Ferroptosis has been recognized as a potential treatment for various cancers. Still, in the complex tumor microenvironment, the communication between cancer cells and tumor-associated macrophages (TAMs) plays a crucial role in tumorigenesis and progression. In this work, scanning electrochemical microscopy (SECM) has been combined with microfluidic devices to enable on-chip cell coculture and in situ investigation of the communication between triple-negative breast cancer cells (TNBCs) and TAMs in ferroptosis. In the coculture system, TNBCs and TAMs were used as responding cells and signaling cells, respectively. By in situ monitoring the changes of key parameters (ROS, glutathione (GSH), and cell membrane permeability) in Erastin-induced ferroptosis, it was found that TAMs partially restored the reduced GSH efflux, increased ROS release, and impaired cell membrane barrier in TNBCs, indicating that TAMs can suppress TNBC ferroptosis. Mechanistically, TNBCs could promote M2 macrophage polarization, and M2-TAMs achieved suppression of TNBCs ferroptosis through the STAT3-related signaling pathway. After inhibition of STAT3, increased ROS release and membrane permeability as well as decreased GSH efflux of TNBCs were in situ monitored by SECM, demonstrating the intercellular communication mechanism in ferroptosis. Therefore, this work provides a potential strategy of targeting TAMs for ferroptosis-based TNBC therapy.
Collapse
Affiliation(s)
- Yuqi Lu
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Yuxi Ge
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Feng Tu
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Xin Li
- School of Physics and Electronic Science, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Ping Geng
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Fan Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Qingjiang Wang
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| |
Collapse
|
19
|
Xu S, Lei L, Yang Z, Wang Y, Du S, Zhao Q, Huang X, Cao S, Wu R, Wang Y, Yan Q, Wen Y. Cytolethal distending toxin from Glaesserella parasuis induces ferroptosis in porcine alveolar macrophages and mice. Vet Res 2025; 56:92. [PMID: 40281583 PMCID: PMC12023646 DOI: 10.1186/s13567-025-01520-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/09/2025] [Indexed: 04/29/2025] Open
Abstract
Glaesserella parasuis cytolethal distending toxin (GpCDT) is a bacterial genotoxin whose main action is to activate DNA damage responses, induce cell cycle arrest, and induce the apoptosis of host cells. In our previous studies, we reported that cells incubated with GpCDT exhibited changes in the expression of ferroptosis-related proteins; thus, we hypothesized that, in addition to apoptosis, GpCDT may also cause ferroptosis, a novel mode of cell death. Here, we observed that treatment of 3D4/21 cells with GpCDT resulted in cytoplasmic iron overload, depletion of GSH (reduced glutathione), and overproduction of reactive oxygen species (ROS) and malondialdehyde (MDA), indicating that GpCDT disrupted iron metabolism and redox homeostasis in these cells. These phenomena were counteracted by the specific ferroptosis inhibitor ferrostatin-1 and the iron chelator deferoxamine mesylate. In vitro infection with the Glaesserella parasuis field isolate strain SC1401 (CDT positive) induced changes in the expression of ferroptosis biomarkers and proteins. Infection of C57BL/6 mice yielded similar results. Our results suggest that ferroptosis may play a substantial role in GpCDT-induced cellular injury.
Collapse
Affiliation(s)
- Shiyu Xu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Li Lei
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Zhen Yang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- National Center of Technology Innovation for Pigs, Chongqing, 402460, China
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China
| | - Yu Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Senyan Du
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Qin Zhao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, 61130, China
| | - Xiaobo Huang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, 61130, China
| | - Sanjie Cao
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, 61130, China
| | - Rui Wu
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yiping Wang
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Qigui Yan
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yiping Wen
- Research Center for Swine Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
- Sichuan Science-Observation Experimental Station of Veterinary Drugs and Veterinary Biotechnology, Ministry of Agriculture and Rural Affairs, Chengdu, 61130, China.
| |
Collapse
|
20
|
Liu C, Liu X, Wang Y, Yu H, Li Q, Zheng Y, Fu Y, Yao G, Sun L. Mesenchymal stromal cells reduce ferroptosis of podocytes by activating the Nrf2/HO-1/GPX4 pathway in lupus nephritis. Int Immunopharmacol 2025; 153:114537. [PMID: 40147265 DOI: 10.1016/j.intimp.2025.114537] [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: 05/20/2024] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Ferroptosis has been reported to be involved in the occurrence and development of various kidney diseases. Emerging evidence suggests that ferroptosis also plays a critical role in systemic lupus erythematosus (SLE) and lupus nephritis (LN), contributing to podocyte injury and renal dysfunction. Mesenchymal stromal cells (MSCs) have become an attractive option for podocyte injury repairing in LN. The aim of this research was to determine whether MSCs regulate ferroptosis of podocytes in LN. METHODS MSCs were injected into female MRL/lpr mice via tail vein. The symptoms of LN and the detection of ferroptosis-related biomarkers in podocytes were detected. In vitro validation was conducted by mouse podocyte cell line MPC-5. RESULTS The occurrence of ferroptosis and involvement of Nrf2/heme oxygenase-1 (HO-1) signaling pathway in podocytes were observed. We found increased expression of the podocyte marker, Wilm's tumor 1 (WT-1) and synaptopodin, following the improvement of lupus-like symptoms after MSC transplantation in MRL/lpr mice. The expression of ferroptosis-related protein glutathione peroxidase 4 (GPX4) and long chain acyl-CoA synthetase 4 (ACSL4) were elevated in renal, along with the Nrf2 and HO-1 activity enhancement. In vitro, MSC treatment maintain a stabilization of podocyte actin stress fibers, leading to an improvement of cell viability. Furthermore, our results showed that puromycin aminonucleoside (PAN) induce accumulation of cellular lipid reactive oxygen species (ROS) and glutathione depletion, and the expression of Nrf2, HO-1 and GPX4 were all downregulated whereas the expression of ACSL4 was upregulated. However, these effects were reversed by MSCs and ferroptosis inhibitor ferrastatin-1 (Fer-1). The promotion of Nrf2 nuclear translocation was observed after the treatment with MSCs. CONCLUSION Ferroptosis activation is involved in the development of LN. MSCs could ameliorate podocyte injury in LN by inhibiting ferroptosis through the Nrf2/HO-1/GPX4 pathway, which will provide novel potential therapeutic targets for LN.
Collapse
Affiliation(s)
- Chang Liu
- Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Xuanqi Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yujiao Wang
- Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Honghong Yu
- Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Qi Li
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuanyuan Zheng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yao Fu
- Department of Pathology, Affiliated Drum Tower Hospital, Medical School of Nanjing University.
| | - Genhong Yao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China.
| | - Lingyun Sun
- Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China; Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China; Department of Rheumatology and Immunology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
21
|
Ali A, Ali SL, Ullah W, Khan A. Gene Expression Profiling Identifies CAV1, CD44, and TFRC as Potential Diagnostic Markers and Therapeutic Targets for Multiple Myeloma. Cell Biochem Biophys 2025:10.1007/s12013-025-01743-0. [PMID: 40246772 DOI: 10.1007/s12013-025-01743-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/19/2025]
Abstract
Multiple myeloma (MM) is a highly malignant hematological tumor with a low overall survival rate, making the identification of innovative prognostic markers essential due to its complex and heterogeneous nature. Ferroptosis, an iron-dependent form of cell death driven by lipid peroxidation, is now recognized as crucial in tumor development and progression. Consequently, ferroptosis-related genes (FRGs) are emerging as promising therapeutic targets and prognostic indicators. However, the specific roles and predictive value of FRGs in MM still remain unclear. The current study was therefore conceived to examine the possible involvement of FRGs in MM. FRGs data was obtained from the FerrDb resource. The datasets GSE133346 and GSE166122, sourced from the Gene Expression Omnibus (GEO), provided gene expression data for both healthy and MM individuals. The differentially expressed-FRGs (DE-FRGs) were identified using the limma and DESeq2 packages in R. Functional pathways were analyzed through Gene Ontology (GO) and KEGG enrichment analyses. The miRWalk database was used for miRNA association and enrichment analysis with hub genes. Prognosis-related genes were evaluated using Kaplan-Meier survival analyses. We identified 1400 differentially expressed genes and cross-referenced them with FRGs, ultimately selecting 17 as DE-FRGs or hub genes. GO analysis revealed that the primary enriched functions of these hub genes are sister chromatid segregation, condensed chromosome centromeric region, C-C chemokine receptor activity, and C-C chemokine binding. KEGG pathway analysis showed that these overlapped genes were enriched in several pathways, including cell cycle, viral protein interaction with cytokine and cytokine receptor, as well as breast and prostate cancers involved pathways. Furthermore, significant enrichment was observed in glycolysis, gluconeogenesis, and the citrate cycle pathways based on miRNAs association with the candidate genes. The CAV1, CD44, TFRC, DPP4, and GJA1 are identified as top five significant hub DE-FRGs based on protein-protein interaction (PPI) analysis from multiple resources. Survival analysis eventually identified CAV1, CD44, and TFRC as the top-ranked DE-FRGs associated with overall survival, underscoring their crucial role in MM. This study identifies CAV1, CD44, and TFRC as key FRGs associated with the prognosis of MM, suggesting their potential as valuable prognostic markers and therapeutic targets to improve patient outcomes.
Collapse
Affiliation(s)
- Awais Ali
- Department of Biochemistry, Abdul Wali Khan University Mardan (AWKUM), Mardan, Pakistan
| | - Syed Luqman Ali
- Department of Biochemistry, Abdul Wali Khan University Mardan (AWKUM), Mardan, Pakistan
| | - Waseef Ullah
- Department of Biochemistry, Abdul Wali Khan University Mardan (AWKUM), Mardan, Pakistan
| | - Asifullah Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan (AWKUM), Mardan, Pakistan.
| |
Collapse
|
22
|
Yang H, Xiao G, Wang D, Xiong T, Wang J, Jing X, Xiong B, Xie J, Liu B, She Q. Inhibition of HMOX1 alleviates diabetic cardiomyopathy by targeting ferroptosis. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40235314 DOI: 10.3724/abbs.2024232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025] Open
Abstract
Diabetic cardiomyopathy (DCM) is an important complication of chronic diabetes mellitus. However, its pathologic process and pathogenesis have not been fully elucidated. This study aims to investigate the role of ferroptosis in DCM and clarify the effect of heme oxygenase-1 (HMOX1) on DCM by targeting ferroptosis. In vivo, an animal model of DCM is established by subjecting mice to a high-fat diet (HFD) combined with low-dose streptozotocin (STZ) injection. We induce an in vitro DCM model by exposing H9C2 cells to high glucose and palmitic acid. Transcriptome sequencing reveals that the differentially expressed genes (DEGs) are enriched primarily in fatty acid metabolism and mitochondrial fatty acid β-oxidation, which are closely related to ferroptosis. The experimental results show that the diabetic microenvironment induces ferroptosis both in vivo and in vitro. Western blot analysis reveals the decreased expressions of the antioxidant proteins GPX4, SLC7A11 and ferritin in the DCM group. However, qPCR demonstrates the elevated expressions of the ferroptosis markers PTGS2 and ACSL4. Biochemical indicators further support the occurrence of ferroptosis, with increased levels of malondialdehyde (MDA) and lactate dehydrogenase (LDH), along with decreased level of glutathione (GSH). In vitro, intervention with high glucose and palmitic acid in H9C2 cells results in ferroptosis, which is reversed by ferrostatin-1 (Fer-1). Results show the elevated expression of HMOX1 in DCM. Moreover, knockdown of HMOX1 ameliorates ferroptosis, thereby alleviating diabetic cardiomyopathy by reducing cardiac fibrosis and improving cardiac function. Our study elucidates the role of HMXO1 in DCM pathogenesis and provides a potential therapeutic strategy for clinical treatment.
Collapse
Affiliation(s)
- Huiping Yang
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Gongyi Xiao
- Department of Orthopedic Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
- Department of Orthopedic Surgery, Chonggang General Hospital, Chongqing 400000, China
| | - Dinghui Wang
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Tianhua Xiong
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jing Wang
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiaodong Jing
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Bingquan Xiong
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Junmei Xie
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Bin Liu
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qiang She
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
23
|
Elhami A, Ghasemzadeh S, Emrahoglu S, Ghasemzadeh N, Mivehchi H, Beykoylu M, Ashrafpour M, Ayoubi S, Tabari MAK. Targeting ferroptosis: a novel pathway in oral, oropharyngeal, hypopharyngeal, and laryngeal cancers. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04142-7. [PMID: 40227310 DOI: 10.1007/s00210-025-04142-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
Malignancies of the oral cavity, oropharynx, hypopharynx, and larynx rank as the seventh most prevalent cancers globally, characterized by high morbidity and mortality. Despite advancements in conventional therapies, these cancers often demonstrate recurrence and treatment resistance. This review investigates ferroptosis, an iron-dependent regulated cell death mechanism, as a novel therapeutic target to overcome resistance and recurrence in these cancers. A narrative review study was conducted using online databases, including PubMed, Google Scholar, Scopus, and Web of Science. The search incorporated keywords such as "ferroptosis", "oral squamous cell carcinoma", "oropharyngeal cancer", "hypopharyngeal cancer", "laryngeal cancer", "iron metabolism", and "lipid peroxidation". Studies focusing on molecular mechanisms, ferroptosis regulation, and therapeutic applications were included. Key findings highlighted the involvement of genes like CA9, CAV1, and SLC7 A11 in oral squamous cell carcinoma (OSCC), contributing to progression and resistance. Ferroptosis inducers such as resveratrol and quercetin effectively promoted ferroptosis in OSCC by targeting pathways like p53/SLC7 A11. In hypopharyngeal and oropharyngeal cancers, agents like ascorbic acid and RSL3 enhanced lipid peroxidation, while laryngeal cancers showed resistance through molecules like SLC3 A2 and KPNA2, which could be counteracted with targeted therapies. Nanotechnology-based approaches, including photodynamic therapy and nanofiber membranes, offer potential for localized and effective ferroptosis induction. Ferroptosis holds promise as a therapeutic strategy for treating head and neck cancers by addressing treatment resistance and recurrence. Future research should focus on optimizing combination therapies, understanding molecular heterogeneity, and translating preclinical findings into clinical applications to improve patient outcomes.
Collapse
Affiliation(s)
- Anis Elhami
- Faculty of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | | | - Sahand Emrahoglu
- School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Nasim Ghasemzadeh
- School of Mathematics and Natural Science, University of Dallas at Texas, Richardson, TX, USA
| | - Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - Saminalsadat Ayoubi
- School of Dental Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
24
|
Anash M, Maparu K, Singh S. Unraveling cell death mechanisms in traumatic brain injury: dynamic roles of ferroptosis and necroptosis. Mol Biol Rep 2025; 52:381. [PMID: 40208458 DOI: 10.1007/s11033-025-10489-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Traumatic brain injury (TBI) remains a major cause of mortality and long-term disability worldwide, with ferroptosis and necroptosis emerging as key drivers of secondary neuronal damage. Ferroptosis, characterized by iron-dependent lipid peroxidation and mitochondrial dysfunction, exacerbates oxidative stress and neuronal cell death. In parallel, necroptosis, mediated by receptor-interacting protein kinases (RIPK1 and RIPK3), amplifies inflammation through membrane rupture and the release of cellular components. Mitochondrial dynamics, involving fission and fusion processes, play a dual role in regulating these pathways. While mitochondrial fusion preserves cellular integrity and reduces oxidative stress, excessive mitochondrial fission driven by dynamin-related protein 1 (DRP1) accelerates necroptotic signaling and neuronal injury. This intricate interplay between ferroptosis, necroptosis, and mitochondrial dynamics highlights potential therapeutic targets. Modulating these pathways through tailored interventions could reduce neuronal damage, mitigate neuroinflammation, and improve functional outcomes in TBI patients. Advancing our understanding of these mechanisms is essential for developing precision therapies that address the complex pathology of traumatic brain injury.
Collapse
Affiliation(s)
- Mohd Anash
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Kousik Maparu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
25
|
Song W, Sun P, Zhao T, Zang Y, Dong P, Tang Q, Chen W, Chen W, Wang Z, Zhang Q, Wang Y, Yin C, Yu M. Unveiling the therapeutic potential of ferroptosis in lung cancer: a comprehensive bibliometric analysis and future therapeutic insights. Discov Oncol 2025; 16:508. [PMID: 40208519 PMCID: PMC11985706 DOI: 10.1007/s12672-025-02234-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Lung cancer remains the leading cause of cancer-related deaths worldwide, with increasing attention being given to novel therapeutic strategies that target the mechanisms underlying tumor growth and drug resistance. Among these, ferroptosis, a regulated cell death driven by iron-dependent lipid peroxidation, has become a key focus in cancer research. Despite extensive research, the exact role of ferroptosis in lung cancer progression and treatment remains unclear, especially regarding its interaction with immune cells and the tumor microenvironment. OBJECTIVE AND METHODS To address these limitations, this study utilizes a comprehensive bibliometric analysis to explore the current landscape of ferroptosis research in lung cancer. We collected data from the Web of Science Core Collection, covering articles published between 2015 and 2025, and analyzed them using advanced tools such as VOSviewer and CiteSpace. RESULTS This study uses a comprehensive bibliometric analysis to uncover key trends and emerging areas related to lung cancer in ferroptosis research. Recently, the focus has shifted from basic mechanisms to clinical applications, particularly in developing GPX4-targeted therapies and combination treatments. With increasing international collaboration, the United States and China have become key players. Interdisciplinary research, especially on ferroptosis and the cancer-immune system, offers new insights into its role in the tumor microenvironment and immunotherapy. Ferroptosis shows excellent promise in overcoming drug resistance by regulating iron-dependent lipid peroxidation and enhancing treatment efficacy. Future research should focus on ferroptosis' clinical translation, particularly in personalized medicine and overcoming resistance, offering broad prospects for lung cancer treatment. CONCLUSION This paper provides valuable insights into the trends, key contributors, and emerging frontiers of ferroptosis research in lung cancer. It identifies important developments that can serve as a foundation for translating ferroptosis-based therapies into clinical practice, particularly to address drug resistance in lung cancer.
Collapse
Affiliation(s)
- Wenhuan Song
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
- Binzhou Medical University, Binzhou, China
| | - Peipei Sun
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Tongzhen Zhao
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Yunxue Zang
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Pengpeng Dong
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Qi Tang
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China
| | - Wenyu Chen
- Binzhou Medical University, Binzhou, China
| | - Wenyi Chen
- Binzhou Medical University, Binzhou, China
| | | | | | | | - Chunhui Yin
- Shandong Academy of Medical Sciences (SDAMS), Jinan, China
| | - Mingkun Yu
- Binzhou Medical College Affiliated Traditional Chinese Medicine Hospital, Binzhou, China.
- Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
26
|
Prabhune NM, Ameen B, Prabhu S. Therapeutic potential of synthetic and natural iron chelators against ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3527-3555. [PMID: 39601820 DOI: 10.1007/s00210-024-03640-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Ferroptosis, a regulated form of cell death, is characterized by iron accumulation that results in the production of reactive oxygen species. This further causes lipid peroxidation and damage to the cellular components, eventually culminating into oxidative stress. Recent studies have highlighted the pivotal role of ferroptosis in the pathophysiological development and progression of various diseases such as β-thalassemia, hemochromatosis, and neurodegenerative disorders like AD and PD. Extensive efforts are in progress to understand the molecular mechanisms governing the role of ferroptosis in these conditions, and chelation therapy stands out as a potential approach to mitigate ferroptosis and its related implications in their development. There are currently both synthetic and natural iron chelators that are being researched for their potential as ferroptosis inhibitors. While synthetic chelators are relatively well-established and studied, their short plasma half-life and toxic side effects necessitate the exploration and identification of natural products that can act as efficient and safe iron chelators. In this review, we comprehensively discuss both synthetic and natural iron chelators as potential therapeutic strategies against ferroptosis-induced pathologies.
Collapse
Affiliation(s)
- Nupura Manish Prabhune
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Bilal Ameen
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sudharshan Prabhu
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
27
|
Bakar-Ates F, Ozkan E. Synergistic ferroptosis in triple-negative breast cancer cells: Paclitaxel in combination with Erastin induced oxidative stress and Ferroportin-1 modulation in MDA-MB-231 cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3985-3994. [PMID: 39392483 DOI: 10.1007/s00210-024-03523-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Ferroptosis is an important regulated cell death mechanism characterized by iron-dependent lipid peroxidation and oxidative stress. In this study, we examined the ferroptosis-inducing effect of the combined use of Paclitaxel, a microtubule-stabilizing agent, and Erastin, a ferroptosis inducer, in breast cancer cells. In this context, the combination of the compounds in question was applied to the cells and the presence of a synergistic effect was determined by calculating the combination index. Glutathione (GSH) levels and glutathione peroxidase (GPX) activity were determined by commercial assay kits, and the effect of the compounds on lipid peroxidation was determined by measurement of malondialdehyde (MDA) levels. Additionally, the effect of combination treatment on ferroptotic protein expression was determined by western blot. Our findings revealed that the combination treatment caused a significant change in mitochondrial function by causing an increase in the depolarized/viable cell population. Additionally, there was a significant increase in intracellular reactive oxygen species (ROS) levels compared to single applications of the compounds. The significant increase observed in malondialdehyde (MDA) levels revealed that the combination treatment increased lipid peroxidation. Moreover, intracellular GSH levels and glutathione peroxidase (GPX) activity significantly decreased by Paclitaxel-Erastin combination. The expression of ferroptosis-regulating proteins was significantly downregulated. The findings showed that the Paclitaxel-Erastin combination synergistically contributed to the accumulation of lipid reactive oxygen species and induced the ferroptotic cell death pathway in breast cancer cells.
Collapse
Affiliation(s)
- Filiz Bakar-Ates
- Faculty of Pharmacy, Department of Biochemistry, Ankara University, Anadolu, Ankara, 06560, Turkey.
| | - Erva Ozkan
- Faculty of Pharmacy, Department of Biochemistry, Ankara Medipol University, Altindag, Ankara, 06050, Turkey
| |
Collapse
|
28
|
Li P, Chu D, Ding G, Qin D, Bu Y, Tian B. IGF2BP3 suppresses ferroptosis in lung adenocarcinoma by m6A-dependent regulation of TFAP2A to transcriptionally activate SLC7A11/GPX4. Mol Cell Biochem 2025; 480:2361-2375. [PMID: 39026029 DOI: 10.1007/s11010-024-05068-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/30/2024] [Indexed: 07/20/2024]
Abstract
Ferroptosis is recently discovered as an important player in the initiation, proliferation, and progression of human tumors. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) has been reported as an oncogene in multiple types of cancers, including lung adenocarcinoma (LUAD). However, little research has been designed to investigate the regulation of IGF2BP3 on ferroptosis in LUAD. qRT-PCR and western blot were used to measure the mRNA and protein expression of IGF2BP3 and transcription factor AP-2 alpha (TFAP2A). CCK-8 assay was performed to determine cell viability. DCFH-DA and C11-BODIPY staining were used to detect the levels of intracellular reactive oxygen species (ROS) and lipid ROS. The corresponding assay kits were used to analyze the levels of malondialdehyde (MDA) and glutathione (GSH). SRAMP website and m6A RNA immunoprecipitation (Me-RIP) were used to predict and confirm the m6A modification of TFAP2A. RIP experiments were conducted to confirm the binding of IGF2BP3 and TFAP2A. RNA stability assay was performed using actinomycin D. Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter experiments were performed to confirm the interaction between TFAP2A and cystine/glutamate antiporter solute carrier family 7 member 11 (SLC7A11) or glutathione peroxidase 4 (GPX4). Mice xenotransplant model was also constructed to explore the effect of IGF2BP3 on LUAD tumor growth and ferroptosis. IGF2BP3 and TFAP2A were both highly expressed in LUAD. IGF2BP3 or TFAP2A knockdown induced ferroptosis by aggravating erastin-induced cell viability suppression, increasing the production of intracellular ROS, lipid ROS, and MDA, and decreasing GSH synthesis, GSH/GSSG ratio, and cystine uptake. Mechanistically, IGF2BP3 stabilized TFAP2A expression via m6A modification. Moreover, sh-IGF2BP3-mediated ferroptosis was significantly abated by TFAP2A overexpression. Furthermore, TFAP2A binds to the promoters of SLC7A11 and GPX4 to promote their transcription. Also, IGF2BP3 depletion suppressed LUAD tumor growth by inducing ferroptosis in mice. IGF2BP3 suppresses ferroptosis in LUAD by m6A-dependent regulation of TFAP2A to promote the transcription of SLC7A11 and GPX4. Our findings suggest that targeting IGF2BP3/TFAP2A/SLC7A11/GPX4 axis might be a potential therapeutic choice to increase ferroptosis sensitivity in LUAD.
Collapse
Affiliation(s)
- Pengpeng Li
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Dan Chu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, China.
| | - Guangcheng Ding
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Dehua Qin
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Yajing Bu
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China
| | - Bi Tian
- Tumor Treatment Center, The Fifth Affiliated Hospital of Zhengzhou University, No. 3 Kangfuqian Street, Erqi District, Zhengzhou, 450052, China.
| |
Collapse
|
29
|
Hao X, Qian X, Xie C, Wang Z, Wang X, Ji Y, Zhang X, Li Q, Wan B, Cui H, Wang L, Yang N, Qiao L, Yu H, Han F, Zhuang H, Zhou J. CircMFN2/miR-361-3p/ELK1 feedback loop promotes glutaminolysis and the progression of hepatocellular carcinoma. Cancer Lett 2025; 614:217473. [PMID: 39933635 DOI: 10.1016/j.canlet.2025.217473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/23/2024] [Accepted: 01/17/2025] [Indexed: 02/13/2025]
Abstract
Current evidence indicates that circRNAs are involved in the development of multiple malignancies including hepatocellular carcinoma (HCC). However, the specific functions of circRNAs in HCC metabolism and progression and their underlying regulatory mechanisms remain unclear. We have identified a novel circRNA circMFN2, by bioinformatics analysis of circRNA microarray data from the GEO database. The levels of circMFN2 were assessed in HCC cell lines and tissues, and its clinical relevance was assessed. The effect of circMFN2 on HCC cells was evaluated in vitro and in vivo. The effect of ELK1 on glutaminolysis and HCC progression was also explored. Patients with HCC and high circMFN2 expression exhibited worse survival outcomes. Functionally, downregulation of circMFN2 repressed the proliferation, invasion, and migration of HCC cells in vitro, whereas ectopic expression of circMFN2 had the opposite effects. The effects of tumor enhancement by circMFN2 on HCC were confirmed by in vivo experiments. Mechanistically, circMFN2 acted as a sponge for miR-361-3p, leading to the upregulation of its target ELK1, whereas ELK1 was enriched in the MFN2 promoter to enhance the transcription and expression of MFN2, indirectly leading to the upregulation of circMFN2. Additionally, we found that circMFN2 promotes glutaminolysis in HCC by increasing ELK1 phosphorylation. We concluded that circMFN2 facilitates HCC progression via a circMFN2/miR-361-3p/ELK1 feedback loop, which promotes glutaminolysis mediated by the upregulation of phosphorylated ELK1. Therefore, circMFN2 not only serves as a potential prognostic indicator, but it could also serve as a therapeutic target for HCC. Further studies are warranted.
Collapse
Affiliation(s)
- Xiaopei Hao
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiangjun Qian
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Chenxi Xie
- Department of Hepatobiliary Surgery, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengzheng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Xiaoqian Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yang Ji
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing, China
| | - Xiaokai Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Qingjun Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Baishun Wan
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Hong Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Li Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Nanmu Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney at Westmead Hospital, Westmead, NSW, 2145, Australia.
| | - Haibo Yu
- Department of Hepatobiliary Surgery, People's Hospital of Zhengzhou University, Zhengzhou, China.
| | - Feng Han
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China.
| | - Hao Zhuang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China.
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China.
| |
Collapse
|
30
|
Ma Z, Wang Y, Zhang X, Ding S, Fan J, Li T, Xiao X, Li J. Curculigoside exhibits multiple therapeutic efficacy to induce apoptosis and ferroptosis in osteosarcoma via modulation of ROS and tumor microenvironment. Tissue Cell 2025; 93:102745. [PMID: 39864205 DOI: 10.1016/j.tice.2025.102745] [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: 07/22/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/28/2025]
Abstract
OBJECTIVE Patients with osteosarcoma (OS) exhibit metastasis upon diagnosis, and the condition frequently acquires resistance to traditional chemotherapy treatments, failing the therapy. The objective of this research was to examine the impact of curculigoside (Cur), a key phenolic compound discovered in the rhizome of C. orchioides Gaertn, on OS cells and the surrounding tumor environment. METHODS We assessed the impact of curculigoside on tumor inhibition in four osteosarcoma cell lines and mice tumor xenograft models using various techniques including cell viability assay, wound healing assay, cell apoptosis analysis, immunofluorescent staining, and IHC. Moreover, we created a mini-PDX model by utilizing freshly obtained primary OS cells from surgically removed OS tissues to evaluate the possible clinical use of Cur. RESULT The results of our study show that Cur triggers cell death in OS cells and enhances the maturation of RAW264.7 cells. By effectively inhibiting the growth of OS cells, these actions mechanistically trigger the catastrophic buildup of unbound iron and uncontrolled lipid peroxidation, ultimately resulting in ferroptosis. Moreover, additional validation of Cur's substantial antineoplastic impact is obtained through in vivo experiments employing xenograft and mini-PDX models. CONCLUSIONS To sum up, this research is the initial one to exhibit the anti-tumor effects of Cur on OS using various methods, indicating that Cur shows potential as a viable approach for treating OS.
Collapse
Affiliation(s)
- Ziyang Ma
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an 710032, China
| | - Yirong Wang
- Department of Endodontics, School of Stomatology, The Air Force Military Medical University, Xi'an 710032, China
| | - Xiaoyu Zhang
- Affiliated Medical College, Yan'an University, Xi'an 716000, China
| | - Shi Ding
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an 710032, China
| | - Jian Fan
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an 710032, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, 8 Changjiang Avenue, Tianjin 300100, China.
| | - Xin Xiao
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an 710032, China.
| | - Jing Li
- Department of Orthopedics, Xijing Hospital, The Air Force Medical University, Xi'an 710032, China.
| |
Collapse
|
31
|
Yin Y, Mu F, Zhang L, Zhao J, Gong R, Yin Y, Zheng L, Du Y, Jin F, Wang J. Wedelolactone activates the PI3K/AKT/NRF2 and SLC7A11/GPX4 signalling pathways to alleviate oxidative stress and ferroptosis and improve sepsis-induced liver injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119557. [PMID: 40010556 DOI: 10.1016/j.jep.2025.119557] [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: 12/20/2024] [Revised: 02/11/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
INTRODUCTION Sepsis-induced liver injury (SILI) is a severe complication of sepsis. Wedelolactone (WEL) can be used to treat liver diseases. However, its therapeutic mechanisms and efficacy in SILI remain unclear. To investigate the therapeutic effects of WEL on SILI and its potential mechanisms of action through in vitro and in vivo experiments. METHODS A SILI model based on lipopolysaccharide (LPS), and AML12 cells were treated with different concentrations of WEL, LY294002 and ML385. The SILI model was established by caecal ligation and puncture (CLP). C57BL/6 mice were administered WEL and biphenyl diester for seven consecutive days, and CLP was then performed 1 h later. Blood and liver tissue were collected 24 h later for subsequent analysis. HE staining, liver function index, oxidative stress index, JC-1 staining, transmission electron microscopy, immunofluorescence staining, Western blot, and inflammatory cytokines were used to detect oxidative stress and ferroptosis-related markers. RESULTS The in vivo experiments showed that WEL treatment reduced the pathological damage of the liver and decreased ALT and AST, MMP and ROS (the product of iron and lipid peroxidation) and inflammatory factors. WEL also decreased hepatocyte viability in vitro. Inhibition of NRF2 can lead to exacerbation of SILI. The expressions of P-PI3K and P-AKT were up-regulated while HO-1, GPX4, NRF2, and SLC7A11 were down-regulated in vitro and in vivo. CONCLUSIONS Ferroptosis and oxidative stress are pivotal in SILI. WEL mitigates SILI by inhibiting ferroptosis and oxidative stress, primarily through the PI3K/AKT/NRF2 and SLC7A11/GPX4 signalling pathways, thus suggesting a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yanping Yin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China; College of Life Science, Northwest University, Xi'an, 710069, China
| | - Fei Mu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lulu Zhang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinyi Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Gong
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yanli Yin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lingling Zheng
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yang Du
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Fuxing Jin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
32
|
Zhan T, Liu Y, Duan S, Lu C, Jia H, Jin M, Li J, Du X, Sun S, Li Y, Zhang J. Targeting HCG18 counteracts ferroptosis resistance via blocking the miR-30a-5p/RRM2/GSS pathway in hepatocellular carcinoma. Int J Biol Sci 2025; 21:2550-2567. [PMID: 40303288 PMCID: PMC12035896 DOI: 10.7150/ijbs.104127] [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: 09/24/2024] [Accepted: 03/07/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Finding effective strategies and novel targets for reversing drug resistance is one of the major frontiers in hepatocellular carcinoma (HCC) research. Ferroptosis is participate in the malignant progression and drug resistance of HCC. However, the underlying molecular mechanisms remail largely uninvestigated. Methods: HCC cell lines and xenografted nude mice were used as experimental models. Biological functions were investigated by various molecular biology experiments. An HCC population was used to reveal clinical significance. Results: In our study, HCG18 and RRM2 was found to be associated with unfavorable prognosis. HCG18 regulates RRM2 expression through competitively binding to miR-30a-5p, consequently impacting ferroptosis. RRM2 directly regulated GSS to increase GSH synthesis. The colony formation assay demonstrated that overexpression of HCG18 inhibited erastin-induced cell death. In addition, in vivo experiments have also confirmed that HCG18 can inhibit ferroptosis by regulating the expression of RRM2, thereby promoting HCC proliferation. Conclusion: Our study discovered a novel lncRNA HCG18, as a "switch-like" molecule of the axis of miR-30a-5p/RRM2/GSS, confers resistance to ferroptosis and holds promise as a potential target for ferroptosis-dependent therapy.
Collapse
Affiliation(s)
- Tian Zhan
- Department of General Surgery, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Yawei Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Shuoke Duan
- Department of Gastroenterology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215031, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chen Lu
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, 211100, China
| | - Heng Jia
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Ming Jin
- Department of Gastroenterology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215031, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jie Li
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Xinru Du
- Department of Gastroenterology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215031, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Sizheng Sun
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Yuan Li
- Department of Gastroenterology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215031, China
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jianping Zhang
- Xiamen Humanity Hospital, Fujian Medical University, Fujian, 350122, China
| |
Collapse
|
33
|
Toczylowska B, Skowronska M, Kurkowska-Jastrzebska I, Ruszczynska A, Zieminska E. Serum metabolomics indicates ferroptosis in patients with pantothenate kinase associated neurodegeneration. Sci Rep 2025; 15:9592. [PMID: 40113937 PMCID: PMC11926261 DOI: 10.1038/s41598-025-94838-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Abstract
The core syndrome among NBIA disorders is pantothenate kinase-associated neurodegeneration (PKAN), an autosomal recessive disorder caused by mutations in the PANK2 gene. There is no therapy for PKAN; only symptomatic treatment is available. Our work aimed to identify the mechanisms induced by biochemical disturbances in the cell cycle and identify potential pharmacological targets to improve patient quality of life. Mass spectrometry (MS) (metals) and NMR spectroscopy (hydrophilic and hydrophobic compounds) were used for profile analyses of the sera of 12 PKAN patients and 12 controls to study the compounds involved in PKAN pathomechanisms. We performed ANOVA and multivariate analysis using orthogonal partial least squares discriminant analysis. We have shown for the first time that patients have 100-500-fold greater serum citrate levels than controls do, which may contribute to Fe transport and ferroptosis. Ferroptosis may be indicated by disturbances in the levels of many metals, oxidative stress, disturbances in energy production and neurotransmission or dysfunction of biological membranes. Our findings suggest that ferroptosis could be a primary cause of cell death in PKAN patients. This could be indicated by serum metabolomics.
Collapse
Affiliation(s)
- Beata Toczylowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, PAS, Ks. Trojdena 4 St., 02-109, Warsaw, Poland
| | - Marta Skowronska
- 2nd Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9 St., 02-957, Warsaw, Poland
| | - Iwona Kurkowska-Jastrzebska
- 2nd Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9 St., 02-957, Warsaw, Poland
| | - Anna Ruszczynska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Zwirki I Wigury 101 St., 02-089, Warsaw, Poland
| | - Elzbieta Zieminska
- Mossakowski Medical Research Institute, PAS, A. Pawinskiego 5 St., 02-106, Warsaw, Poland.
| |
Collapse
|
34
|
Shen S, Pan L, Li J, Wang J, Ahmad I, Liu H, Bai Y, Kang B, Yin J, Gao Y, Lu Y, Wang X. The Involvement of Amino Acid Metabolism in the Mechanisms of Salt Tolerance Adaptation in Medicago sativa and Medicago truncatula. PLANTS (BASEL, SWITZERLAND) 2025; 14:929. [PMID: 40265823 PMCID: PMC11945280 DOI: 10.3390/plants14060929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 04/24/2025]
Abstract
Amino acid metabolism constitutes a major metabolic pathway in plants, playing an important role in the modulation of plant responses to stress. In this study, we investigated the amino acid metabolism responses of M. sativa (Medicago sativa L.) and M. truncatula (Medicago truncatula L.) plants under salt stress using transcriptomic and proteomic approaches to elucidate their salt stress tolerance mechanisms in relation to the regulation of amino acid homeostasis. Transcriptome and proteome sequencing followed by Kyoto Gene and Genome Encyclopedia enrichment analysis revealed 34 differentially expressed genes and 45 differentially expressed proteins involved in valine, leucine, and isoleucine degradation, tyrosine metabolism, and glutathione metabolism. Significant differences were observed in the expression of glutathione S-transferase (GST) within the glutathione metabolic pathway between M. sativa and M. truncatula. The induction of valine, leucine, and isoleucine metabolism, aldehyde dehydrogenases (ALDHs), and alanine-glyoxylate aminotransferases (AGXTs), involved in intracellular reactive oxygen species scavenging, also significantly differed under salt stress. Significant differences were identified in the expression of tyrosine decarboxylases (TDCs) involved in tyrosine metabolism, which are responsible for tyramine biosynthesis and can enhance plant tolerance to salt stress. This study delved into the effects of amino acid metabolism on the salt tolerance mechanisms of M. sativa and M. truncatula, which is crucial in guiding the future breeding of salt-tolerant alfalfa varieties.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xiaoshan Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (S.S.); (L.P.); (J.L.); (J.W.); (I.A.); (H.L.); (Y.B.); (B.K.); (J.Y.); (Y.G.); (Y.L.)
| |
Collapse
|
35
|
Zhou J, Wang J, Yang L, Fu T, Li H, Shan Y, Gao H, Xie C, Zhang L, Zhang M, Ma J, Liu L, Fang H, Jiang D, Xu M, Pan Q, Gu S. N6-methyadenosine-modified YWHAE mRNA promotes proliferation and inhibits ferroptosis in hepatoblastoma by mediating SLC7A11 expression. Oncogene 2025:10.1038/s41388-025-03334-y. [PMID: 40074884 DOI: 10.1038/s41388-025-03334-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 02/03/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
Hepatoblastoma (HB) is a rare but predominant liver cancer in children, with few treatment choices in advanced stages. YWHAE is closely related to several human diseases and acts as a molecular scaffold for malignant transformation. However, whether YWHAE promotes HB development remains unknown. Conducting RNA and m6A sequencing on HB tissues, we found that YWHAE was upregulated and modified by N6-methyadenosine. Functionally, YWHAE promoted proliferation and inhibited cell death in HB by in vitro and in vivo studies. Mechanistically, METTL3-dependent m6A modification activated YWHAE mRNA expression, and the m6A reader IGF2BP2 recognized and bound to the m6A site on YWHAE mRNA, thereby enhancing the mRNA stability of YWHAE. Interestingly, RNA sequencing revealed that YWHAE knockdown was involved in regulating ferroptosis of HB cells by mediating SLC7A11 expression. Moreover, knockdown of YWHAE significantly increased the levels of lipid ROS and peroxides in HB cells, promoting the susceptibility of HB cells to ferroptosis. In summary, these findings illuminated the role of YWHAE in HB progression and uncovered its relevance to ferroptosis as a new therapeutic target for HB.
Collapse
Affiliation(s)
- Jiquan Zhou
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Jing Wang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Liyuan Yang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Tingyi Fu
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Yuhua Shan
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Hongxiang Gao
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Chenjie Xie
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Lei Zhang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Min Zhang
- Pediatric Translational Medicine Institute and Pediatric Congenital Heart Disease Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Ji Ma
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Li Liu
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Houshun Fang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Dapeng Jiang
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Min Xu
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China
| | - Qiuhui Pan
- Department of Laboratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
| | - Song Gu
- Department of General Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
- Department of International Medical, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, 1678 Dongfang Road, Shanghai, 200127, China.
| |
Collapse
|
36
|
Scattolin T, Cavarzerani E, Alessi D, Mauceri M, Botter E, Tonon G, Caligiuri I, Repetto O, Kamensek U, Brezar SK, Dalla Pozza M, Palazzolo S, Cemazar M, Canzonieri V, Demitri N, Nolan SP, Gasser G, Visentin F, Rizzolio F. Unlocking the potential of organopalladium complexes for high-grade serous ovarian cancer therapy. Dalton Trans 2025; 54:4685-4696. [PMID: 39967474 DOI: 10.1039/d5dt00194c] [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: 02/20/2025]
Abstract
High-Grade Serous Ovarian Cancer (HGSOC) is the most common and lethal subtype of ovarian cancer, known for its high aggressiveness and extensive genomic alterations. Typically diagnosed at an advanced stage, HGSOC presents formidable challenges in drug therapy. The limited efficacy of standard treatments, development of chemoresistance, scarcity of targeted therapies, and significant tumor heterogeneity render this disease incurable with current treatment options, highlighting the urgent need for novel therapeutic approaches to improve patient outcomes. In this study we report a straightforward and stereoselective synthetic route to novel Pd(II)-vinyl and -butadienyl complexes bearing a wide range of monodentate and bidentate ligands. Most of the synthesized complexes exhibited good to excellent in vitro anticancer activity against ovarian cancer cells. Particularly promising is the water-soluble complex bearing two PTA (1,3,5-triaza-7-phosphaadamantane) ligands and the Pd(II)-butadienyl fragment. This compound combines excellent cytotoxicity towards cancer cells with substantial inactivity towards non-cancerous ones. This derivative was selected for further studies on ex vivo tumor organoids and in vivo mouse models, which demonstrate its remarkable efficacy with surprisingly low collateral toxicity even at high dosages. Moreover, this class of compounds appears to operate through a ferroptotic mechanism, thus representing the first such example for an organopalladium compound.
Collapse
Affiliation(s)
- Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Enrico Cavarzerani
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Dario Alessi
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Matteo Mauceri
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Eleonora Botter
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Giovanni Tonon
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Ombretta Repetto
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Simona Kranjc Brezar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Maria Dalla Pozza
- ChimieParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, Università degli Studi di Trieste, Strada di Fiume 447, Trieste, Italy
| | - Nicola Demitri
- Area Science Park, Elettra-Sincrotrone Trieste, S.S. 14 Km 163.5, Basovizza, 34149, Trieste, Italy
| | - Steven P Nolan
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281, S-3, 9000 Ghent, Belgium
| | - Gilles Gasser
- ChimieParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Fabiano Visentin
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Flavio Rizzolio
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| |
Collapse
|
37
|
Hao X, Wang Y, Hou MJ, Liao L, Yang YX, Wang YH, Zhu BT. Raloxifene Prevents Chemically-Induced Ferroptotic Neuronal Death In Vitro and In Vivo. Mol Neurobiol 2025; 62:3934-3955. [PMID: 39354232 PMCID: PMC11790820 DOI: 10.1007/s12035-024-04497-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/12/2024] [Indexed: 10/03/2024]
Abstract
Ferroptosis, a regulated form of cell death characterized by excessive iron-dependent lipid peroxidation, can be readily induced in cultured cells by chemicals such as erastin and RSL3. Protein disulfide isomerase (PDI) has been identified as an upstream mediator of chemically induced ferroptosis and also a target for ferroptosis protection. In this study, we discovered that raloxifene (RAL), a selective estrogen receptor modulator known for its neuroprotective actions in humans, can effectively inhibit PDI function and provide robust protection against chemically induced ferroptosis in cultured HT22 neuronal cells. Specifically, RAL can bind directly to PDI both in vitro and in intact neuronal cells and inhibit its catalytic activity. Computational modeling analysis reveals that RAL can tightly bind to PDI through forming a hydrogen bond with its His256 residue, and biochemical analysis further shows that when PDI's His256 is mutated to Ala256, RAL loses its inhibition of PDI's catalytic activity. This inhibition of PDI by RAL significantly reduces the dimerization of both the inducible and neuronal nitric oxide synthases and the accumulation of nitric oxide, both of which have recently been shown to play a crucial role in mediating chemically induced ferroptosis through subsequent induction of ROS and lipid-ROS accumulation. In vivo behavioral analysis shows that mice treated with RAL are strongly protected against kainic acid-induced memory deficits and hippocampal neuronal damage. In conclusion, this study demonstrates that RAL is a potent inhibitor of PDI and can effectively prevent chemically induced ferroptosis in hippocampal neurons both in vitro and in vivo. These findings offer a novel estrogen receptor-independent mechanism for RAL's neuroprotective actions in animal models and humans.
Collapse
Affiliation(s)
- Xiangyu Hao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Yifan Wang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Ming-Jie Hou
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Lixi Liao
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Yong Xiao Yang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China
| | - Ying-Hua Wang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong (Shenzhen), 2001 Longxiang Road, Longgang District, Shenzhen, 518,172, China.
- Shenzhen Bay Laboratory, Shenzhen, 518,055, China.
| |
Collapse
|
38
|
Yang J, Zhou R, Zhou M, Li X. Atorvastatin inhibits ischemia‒reperfusion-associated renal tubular cell ferroptosis by blocking the PGE2/EP4 signaling pathway. In Vitro Cell Dev Biol Anim 2025; 61:275-287. [PMID: 39920560 DOI: 10.1007/s11626-025-01020-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025]
Abstract
Renal ischemia‒reperfusion (I/R) injury is the main cause of acute kidney injury, and its pathological features are manifested primarily by renal tubular epithelial cell injury. The underlying mechanism involves ferroptosis of renal tubular epithelial cells. Atorvastatin (ATO) regulates ferroptosis, and this study explored its role in I/R-induced ferroptosis of renal tubular epithelial cells. We constructed a renal I/R rat model with bilateral renal pedicles using noninvasive arterial clips and placed HK-2 cells in hypoxia/reoxygenation (H/R) incubators to construct the cell model. The damage to rat kidney tissues and HK-2 cells was assessed using enzyme-linked immunosorbent assay (ELISA), hematoxylin and eosin (H&E) staining, and flow cytometry, and the presence of associated proteins was identified through western blotting. Administering ATO markedly lessened the acute kidney damage caused by I/R, decreased the levels of blood urea nitrogen (BUN) and creatinine (CRE), and prevented apoptosis in renal tubular epithelial cells. Treatment with ATO additionally suppressed the production of inflammatory cytokines (TNF-α, IL-1β, and IL-6) and markers linked to ferroptosis (Fe2+, ROS, MDA, ACSL4, and COX2), thereby reducing acute kidney damage associated with I/R. The expression of PGE2 in renal I/R injury is related to the degree of renal injury, and it mainly regulates ferroptosis by binding to EP4. ATO effectively inhibited the expression of PGE2 and EP4. Overall, this study revealed that ATO inhibited ferroptosis of renal tubular epithelial cells by blocking the PGE2/EP4 signaling pathway, thereby alleviating I/R-induced kidney injury.
Collapse
Affiliation(s)
- Jing Yang
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, China
| | - Rongrong Zhou
- Expanded Program On Immunization Department, Yunnan Center for Disease Control and Prevention, Kunming, 650022, China
| | - Mengjiao Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xinghuan Li
- Department of Anesthesiology, Fuwai Yunnan Cardiovascular Hospital, Kunming, 650102, China.
| |
Collapse
|
39
|
Zhai K, Liu G, Cao C, Wang X. Jujuboside B inhibits proliferation and induces apoptosis and ferroptosis in colorectal cancer cells with potential involvement of the MAPK signaling pathway. Oncol Lett 2025; 29:162. [PMID: 39911154 PMCID: PMC11795164 DOI: 10.3892/ol.2025.14908] [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: 05/13/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and life-threatening malignancies worldwide. Jujuboside B (JUB) is a bioactive compound derived from the seeds of Ziziphus jujuba, known for its potential anticancer properties. The present study aimed to investigate the association of JUB with inhibiting the proliferation, apoptosis and ferroptosis of human CRC cells with mitogen-activated protein kinase (MAPK) pathway regulation. First, the human CRC HCT116 cell line was treated with different concentrations of JUB. Subsequently, cell viability was evaluated using MTT assay and colony formation was assessed using a colony formation assay. Flow cytometry was used to detect cell apoptosis and the levels of reactive oxygen species. Western blotting was utilized to assess the expression levels of apoptosis-related proteins, ferroptosis regulators and MAPK pathway-related proteins. In addition, biochemical assay kits were used to evaluate the levels of malondialdehyde, glutathione, total iron and ferrous iron. The results demonstrated that cell viability and colony formation were markedly decreased after JUB treatment, whilst the level of apoptosis was notably increased in a concentration-dependent manner. Using electron microscopy, cells treated with JUB exhibited typical apoptotic bodies, as well as mitochondrial swelling and cristae disruption, further demonstrating JUB-induced cell apoptosis. Western blot analysis indicated that JUB treatment markedly reduced the expression of B-cell lymphoma-2 (Bcl-2) but notably increased the expression of Bcl-2 associated X-protein and cleaved caspase-3. Additionally, JUB induced ferroptosis and inhibited the MAPK signaling pathway in CRC cells. Collectively, the findings of the present study suggest that JUB has the potential to inhibit CRC cell proliferation and induce apoptosis through regulating the MAPK pathway. Therefore, JUB may be a promising therapeutic agent for the treatment of CRC.
Collapse
Affiliation(s)
- Ke Zhai
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Guodong Liu
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Ce Cao
- Department of Gastrointestinal Surgery, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Xiaolong Wang
- Department of Gastrointestinal Surgery, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| |
Collapse
|
40
|
Chen J, Lei KW, Li SY, Li DP, Wang YL, Wang X, Bai X, Huang YL. Dose effects of iron on growth, antioxidant potential, intestinal morphology, and intestinal barrier in yellow-feathered broilers. Poult Sci 2025; 104:104865. [PMID: 39919560 PMCID: PMC11851222 DOI: 10.1016/j.psj.2025.104865] [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: 10/22/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
This experiment was conducted to investigate the dose effects of iron on growth performance, antioxidant function, small intestinal histology, and intestinal barrier of 63-day-old yellow-feathered broilers. A total of 720 1-day-old male yellow-feathered broilers were randomly divided into 9 treatments, with 8 cages per treatment and 10 birds per cage. The Fe supplementation was 0, 20, 40, 60, 80, 160, 320, 640, and 1280 mg/kg, respectively, in the form of FeSO4•7H2O. The results showed that the ADG (P = 0.002) and ADFI (P < 0.001) decreased linearly with increased dietary Fe supplementation. Malondialdehyde (MDA) concentration in plasma (P = 0.001), duodenum (P < 0.001), and jejunum (P < 0.001) were increased linearly as dietary Fe increased. As dietary Fe increased, there was a linear decrease in the villus height and the villus height/crypt depth in the duodenum (P = 0.003; P = 0.001) and jejunum (P = 0.001; P < 0.001). Decreased secretory immunoglobulin A (sIgA) concentration in jejunal mucosa (P < 0.001) was observed with increased dietary Fe concentration. Lower jejunal sIgA concentrations were observed in birds consuming more than 160 mg/kg of Fe (P < 0.001). A quadratic response was found for jejunal diamine oxidase (DAO) activity (P = 0.011) as dietary Fe supplementation was increased. The highest response of DAO in jejunal mucosa was observed for broilers supplemented with 160 mg/kg of Fe. Furthermore, the mRNA expressions of ZO-1 (P < 0.001), occludin (P = 0.004), and claudin-1 (P = 0.007) in jejunal mucosa decreased linearly with increased dietary Fe concentration. Data from the study suggests that there is no need to supplement additional Fe to a corn-soybean-based diet for yellow-feathered broilers based on growth performance, antioxidant potential, small intestinal histology, and intestinal barrier. Chronic iron exposure (≥ 160 mg/kg) can damage the intestinal barrier function, and further increase of Fe supplementation can lead to oxidative stress and even cause growth inhibition for yellow-feathered broilers.
Collapse
Affiliation(s)
- J Chen
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - K W Lei
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - S Y Li
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - D P Li
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - Y L Wang
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - X Wang
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - X Bai
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China
| | - Y L Huang
- Key Laboratory of Animal Science of State Ethnic Affairs Commission, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China; Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, PR China.
| |
Collapse
|
41
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
42
|
Sil R, Chakraborti AS. Major heme proteins hemoglobin and myoglobin with respect to their roles in oxidative stress - a brief review. Front Chem 2025; 13:1543455. [PMID: 40070406 PMCID: PMC11893434 DOI: 10.3389/fchem.2025.1543455] [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: 12/11/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Oxidative stress is considered as the root-cause of different pathological conditions. Transition metals, because of their redox-active states, are capable of free radical generation contributing oxidative stress. Hemoglobin and myoglobin are two major heme proteins, involved in oxygen transport and oxygen storage, respectively. Heme prosthetic group of heme proteins is a good reservoir of iron, the most abundant transition metal in human body. Although iron is tightly bound in the heme pocket of these proteins, it is liberated under specific circumstances yielding free ferrous iron. This active iron can react with H2O2, a secondary metabolite, forming hydroxyl radical via Fenton reaction. Hydroxyl radical is the most harmful free radical among all the reactive oxygen species. It causes oxidative stress by damaging lipid membranes, proteins and nucleic acids, activating inflammatory pathways and altering membrane channels, resulting disease conditions. In this review, we have discussed how heme-irons of hemoglobin and myoglobin can promote oxidative stress under different pathophysiological conditions including metabolic syndrome, diabetes, cardiovascular, neurodegenerative and renal diseases. Understanding the association of heme proteins to oxidative stress may be important for knowing the complications as well as therapeutic management of different pathological conditions.
Collapse
Affiliation(s)
| | - Abhay Sankar Chakraborti
- Department of Biophysics, Molecular Biology and Bioinformatics, University College of Science, University of Calcutta, Kolkata, India
| |
Collapse
|
43
|
Cheng J, Ma X, Tao J, Jiang X, Chen P, Duan X. Neuroprotective effects of ethanol extraction from Rubia yunnanensis Diels on chronic cerebral hypoperfusion: modulation of the System Xc-/GSH/GPX4 axis to alleviate oxidative stress and ferroptosis. Front Pharmacol 2025; 16:1552228. [PMID: 40070574 PMCID: PMC11893507 DOI: 10.3389/fphar.2025.1552228] [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: 12/27/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction Vascular dementia (VD) is a neurodegenerative disease caused by chronic cerebral hypoperfusion (CCH), which considerably impact patients' quality of life. Ethanol extraction from Rubia yunnanensis (RY-A) has gained attention for its potential neuroprotective effects, but its effects and mechanisms of action on CCH are unknown. Methods After 30 days of RY-A gavage treatment in a CCH rat model, its effects were evaluated using the Morris water maze test, cerebral blood flow measurements, and HE staining of the brain. These findings, combined with serum medicinal chemistry, RNA-seq, and metabolomics analyses, revealed the active compounds and mechanisms of RY-A in CCH rats. The results were further validated using assay kits and Western blot techniques. Results RY-A treatment significantly attenuated neurological damage and improved cognitive function in CCH rats. Ultra-high-performance liquid chromatography high-resolution mass spectrometry identified 511 blood-entry compounds of RY-A. RNA-seq and metabolomic analysis showed that RY-A might help to normalize changes in gene and metabolite expression caused by CCH. RY-A induced neuroprotective effects by increasing the production of key proteins involved in ferroptosis inhibition, such as SLC7A11, SLC3A2, GSS, and GPX4, while increasing antioxidant enzyme activities and alleviating oxidative stress. Conclusion RY-A inhibited oxidative stress and ferroptosis by activating the System Xc-/GSH/GPX4 pathway and balancing iron metabolism, thereby attenuating CCH-induced neurological damage and cognitive deficits.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaohua Duan
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
44
|
Li X, Li Y, Xu J, Lu X, Ma S, Sun L, Chang C, Min L, Fan C. Terahertz Wave Desensitizes Ferroptosis by Inhibiting the Binding of Ferric Ions to the Transferrin. ACS NANO 2025; 19:6876-6889. [PMID: 39752147 DOI: 10.1021/acsnano.4c13075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Ferroptosis is a classic type of programmed cell death characterized by iron dependence, which is closely associated with many diseases such as cancer, intestinal ischemic diseases, and nervous system diseases. Transferrin (Tf) is responsible for ferric-ion delivery owing to its natural Fe3+ binding ability and plays a crucial role in ferroptosis. However, Tf is not considered as a classic druggable target for ferroptosis-associated diseases since systemic perturbation of Tf would dramatically disrupt blood iron homeostasis. Here, we reported a nonpharmaceutical, noninvasive, and Tf-targeted electromagnetic intervention technique capable of desensitizing ferroptosis with directivity. First, we revealed that the THz radiation had the ability to significantly decrease binding affinity between the Fe3+ and Tf via molecular dynamics simulations, and the modulation was strongly wavelength-dependent. This result provides theoretical feasibility for the THz modulation-based ferroptosis intervention. Subsequent extracellular and cellular chromogenic activity assays indicated that the THz field at 8.7 μm (i.e., 34.5 THz) inhibited the most Fe3+ bound to the Tf, and the wavelength was in good agreement with the simulated one. Then, functional assays demonstrated that levels of intracellular Fe2+, lipid peroxidation, malondialdehyde (MDA) and cell death were all significantly reduced in cells treated with this 34.5 THz wave. Furthermore, the iron deposition, lipid peroxidation, and MDA in the ferroptosis disease model induced by ischemia-reperfusion injury could be nearly eliminated by the same radiation, validating THz wave-induced desensitization of ferroptosis in vivo. Together, this work provides a preclinical exemplar for electromagnetic irradiation-stimulated desensitization of ferroptosis and predicts an innovative, THz wave-based therapeutic method for ferroptosis-associated diseases in the future.
Collapse
Affiliation(s)
- Xiangji Li
- Department of Gastroenterology, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P. R. China
| | - Yangmei Li
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, P. R. China
| | - Junxuan Xu
- Department of Gastroenterology, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P. R. China
| | - Xinlian Lu
- National Key Laboratory of Human Factors Engineering, China Astronaut Research and Training Center, Beijing 100094, P. R. China
| | - Shixiang Ma
- Department of Retroperitoneal Tumor Surgery, Peking University International Hospital, Beijing 102206, P. R. China
| | - Lan Sun
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P. R. China
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing 100700, P. R. China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing 100071, P. R. China
- School of Physics, Peking University, Beijing 100871, P. R. China
| | - Li Min
- Department of Gastroenterology, State Key Laboratory of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P. R. China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| |
Collapse
|
45
|
Deng RM, Huang G, Wang T, Zhou J. Regulated programmed cell death in sepsis associated acute lung injury: From pathogenesis to therapy. Int Immunopharmacol 2025; 148:114111. [PMID: 39832461 DOI: 10.1016/j.intimp.2025.114111] [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: 09/09/2024] [Revised: 11/28/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Sepsis associated acute lung injury (SALI) is a common complication in patients with severe sepsis and a disease with high morbidity and mortality in ICU patients. The main mechanism of SALI is pulmonary hypoperfusion due to hypotension and shock caused by sepsis, which leads to ischemic necrosis of alveolar endothelial cells and eventually lung failure. At present, SALI therapy mainly includes antibiotic therapy, fluid resuscitation, transfusion products and vasoactive drugs, but these strategies are not satisfactory. Therefore, focusing on the role of different cell death patterns in SALI may help in the search for effective treatments. Understanding the molecular mechanisms of SALI and identifying pathways that inhibit lung cell death are critical to developing effective drug therapies to prevent the progression of SALI. Cell death is controlled by programmed cell death (PCD) pathways, including apoptosis, necroptosis, ferroptosis, pyroptosis and autophagy. There is growing evidence that PCD plays an important role in the pathogenesis of SALI, and inhibitors of various types of PCD represent a promising therapeutic strategy. Therefore, understanding the role and mechanism of PCD in SALI is conducive to our understanding of its pathological mechanism, and is of great significance for the treatment of SALI. In this article, we discuss recent advances in the role of PCD in SALI, show how different signaling pathways (such as NF-κB, PI3K/Akt, mTOR, and Nrf2) regulate PCD to regulate SALI development, and discuss the associations between various types of PCD. The aim is to explore the molecular mechanism behind SALI and to find new targets for SALI therapy.
Collapse
Affiliation(s)
- Rui-Ming Deng
- Department of Anesthesiology, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| | - Guiming Huang
- Department of Anesthesiology, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China
| | - Tingting Wang
- Department of Anaesthesia, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, PR China
| | - Juan Zhou
- Department of Thyroid and Breast Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| |
Collapse
|
46
|
Tong L, Qiu J, Xu Y, Lian S, Xu Y, Wu X. Programmed Cell Death in Rheumatoid Arthritis. J Inflamm Res 2025; 18:2377-2393. [PMID: 39991656 PMCID: PMC11846511 DOI: 10.2147/jir.s499345] [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: 10/05/2024] [Accepted: 02/07/2025] [Indexed: 02/25/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, progressive, systemic autoimmune disease characterised by synovial inflammation, synovial pannus formation and subsequent destruction of articular cartilage and bone. Programmed cell death (PCD), encompassing apoptosis, autophagy, pyroptosis, necroptosis, and ferroptosis, plays a pivotal role in the pathogenesis of RA. An imbalance in PCD causes a variety of immune cells to release large amounts of inflammatory factors and mediators that exacerbate not only chronic synovial inflammation, but also bone and joint damage. The purpose of this article is to review the relevant studies between PCD and RA, with the aim of providing further insights and considerations for a deeper understanding of the pathogenesis of RA and to guide clinical management.
Collapse
Affiliation(s)
- Luyuan Tong
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Jiao Qiu
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Yalin Xu
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Shijing Lian
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Yanqiu Xu
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xiao Wu
- The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| |
Collapse
|
47
|
Wang H, Chen B, Xiao P, Han D, Gao B, Yan Y, Zhao R, Pan T, Zhang J, Zhou M, Lv L, Gao H. Yersiniabactin produced by Escherichia coli promotes intestinal inflammation through lipid peroxidation and ferroptosis. Front Microbiol 2025; 16:1542801. [PMID: 40034497 PMCID: PMC11872927 DOI: 10.3389/fmicb.2025.1542801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Escherichia coli (E. coli), a major foodborne pathogen, poses significant risks to public health by causing gastrointestinal diseases. Among its virulence factors, Yersiniabactin (Ybt), a siderophore, plays a crucial role in iron acquisition and enhancing intestinal colonization. Despite previous studies highlighting E. coli-Ybt's involvement in inflammation, its exact mechanisms remain unclear. This study investigates how Ybt contributes to intestinal inflammation through ferroptosis, using both in vitro and in vivo models. Our findings demonstrate that Ybt promotes oxidative stress, lipid peroxidation, inflammation, and iron accumulation in intestinal epithelial cells, leading to ferroptosis. Mechanistically, Ybt suppresses the Keap1/Nrf2 pathway, amplifying reactive oxygen species (ROS) and activating the TNF/NF-κB pathway, which drives inflammation. Moreover, Ybt induces lipid peroxidation via the arachidonic acid pathway, producing 6-trans-leukotriene B4 (6-transLTB4), which exacerbates inflammation and ferroptosis. Exogenous 6-transLTB4 further intensifies this cascade. Additionally, Ybt disrupts iron efflux by suppressing FPN1 expression, causing excessive intracellular iron accumulation. Using tree shrews as an in vivo model, we confirm that Ybt-induced ferroptosis significantly aggravates intestinal inflammation. These findings underscore the pathogenic role of Ybt in E. coli-induced intestinal injury and highlight ferroptosis as a novel mechanism contributing to gut health disruption. This study provides new insights into the molecular pathways of E. coli infection, with implications for therapeutic strategies targeting ferroptosis in intestinal diseases.
Collapse
Affiliation(s)
- Hao Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Bingxun Chen
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Dongmei Han
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Bin Gao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yulin Yan
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Ru Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Tianling Pan
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Jingsong Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Meng Zhou
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Longbao Lv
- National Resource Center for Non-Human Primates, National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Hong Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
48
|
Ge W, Gao M, Dai Y, Zheng G, Yang L, Zuo W, Tian X. Comprehensive network pharmacology and experimental study to investigate the effect and mechanism of solasonine on breast carcinoma treatment. Cancer Cell Int 2025; 25:49. [PMID: 39962568 PMCID: PMC11834262 DOI: 10.1186/s12935-025-03665-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/26/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Ferroptosis is a therapeutic strategy for breast carcinoma (BC). Solasonine (SS) was linked to ferroptosis as a tumor suppressor. However, whether SS could treat BC by activating ferroptosis and its underlying mechanisms has not been reported. METHODS We obtained the intersection of genes targeting SS and BC disease through network pharmacology. Bioinformatics analysis revealed that the intersection genes were primarily enriched in the extracellular signal-regulated kinase 2/mitogen-activated protein kinase (ERK2/MAPK) signaling pathway. The interaction modes of SS with ERK2 and epidermal growth factor receptor (EGFR) were simulated by molecular docking. We further detected the expressions of ERK2 and p-ERK2 in BC patients and the correlation between ERK2/p-ERK2 and ferroptosis. The effects and mechanism of SS on ferroptosis in BC were validated by mutation plasmids construction, immunohistology, wound healing, transwell assay, and western blotting using in vitro and in vivo models. RESULTS ERK2 and p-ERK2 were up-regulated in BC patients, and the ERK2/p-ERK2 ratio was negatively correlated with ferroptosis. Molecular docking indicated that SS could bind to ERK2 and EGFR to inhibit the activity of the ERK2/MAPK pathway. In vitro and in vivo experiments confirmed that SS induced ferroptosis by inhibiting the ERK2/MAPK pathway, inhibiting proliferation, migration, and invasion of BC cells. CONCLUSION SS could inactivate the ERK2/MAPK pathway, thereby inducing ferroptosis and further inhibiting BC cell proliferation, migration, and invasion. This study clarified the potential mechanism of SS in BC and provided a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Wenkai Ge
- Shandong Provincial Hospital, Shandong University, Jinan, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Min Gao
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yingqi Dai
- Shandong Provincial Third Hospital, Shandong University, Jinan, China
| | - Gang Zheng
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Li Yang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wenshu Zuo
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xingsong Tian
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
49
|
Cilleros-Holgado P, Gómez-Fernández D, Piñero-Pérez R, Romero-Domínguez JM, Reche-López D, Álvarez-Córdoba M, Romero-González A, López-Cabrera A, De Oliveira MC, Rodríguez-Sacristán A, González-Granero S, García-Verdugo JM, Sánchez-Alcázar JA. Polydatin and Nicotinamide Prevent Iron Accumulation and Lipid Peroxidation in Cellular Models of Mitochondrial Diseases. Antioxidants (Basel) 2025; 14:215. [PMID: 40002401 PMCID: PMC11851670 DOI: 10.3390/antiox14020215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Ferroptosis, an iron-dependent form of non-apoptotic cell death, is regulated by a complex network involving lipid metabolism, iron homeostasis, and the oxidative-reductive system, with iron accumulation and lipid peroxidation as key drivers. Mitochondrial dysfunction and ROS overproduction often underlie the pathogenesis of mitochondrial diseases, for which treatment options are limited, emphasizing the need for novel therapies. In this study, we investigated whether polydatin and nicotinamide could reverse ferroptosis-related pathological features in cellular models derived from patients with pathogenic GFM1 variants. Mutant fibroblasts showed increased iron and lipofuscin accumulation, altered expression of iron metabolism-related proteins, elevated lipid peroxidation, and heightened susceptibility to erastin-induced ferroptosis. Treatment with polydatin and nicotinamide effectively corrected these alterations and reduced iron accumulation and lipid peroxidation in induced neurons. Furthermore, chloramphenicol treatment in control cells mimicked the mutant phenotype, suggesting that these pathological changes are linked to the mitochondrial protein synthesis defect characteristic of pathogenic GFM1 variants. Notably, adding vitamin E to the polydatin and nicotinamide co-treatment resulted in a reduction in the minimum effective concentration, suggesting potential benefits of its inclusion. In conclusion, the combination of polydatin, nicotinamide, and vitamin E could represent a promising therapeutic option for patients with mitochondrial disorders caused by pathogenic GFM1 variants.
Collapse
Affiliation(s)
- Paula Cilleros-Holgado
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - David Gómez-Fernández
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Rocío Piñero-Pérez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - José Manuel Romero-Domínguez
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Diana Reche-López
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Mónica Álvarez-Córdoba
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Ana Romero-González
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Alejandra López-Cabrera
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| | - Marta Castro De Oliveira
- Neuropediatria, Neurolinkia, C. Jardín de la Isla, 8, Local 4 y 5, 41014 Sevilla, Spain;
- FEA Pediatría, Centro Universitario Hospitalar de Faro, R. Leão Penedo, 8000-386 Faro, Portugal
| | - Andrés Rodríguez-Sacristán
- Neuropediatría, Servicio de Pediatría, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain;
- Departamento de Farmacología, Radiología y Pediatría de la Facultad de Medicina de la Universidad de Sevilla, 41009 Sevilla, Spain
| | - Susana González-Granero
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46980 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia and CIBERNED-ISCIII, 46980 Valencia, Spain; (S.G.-G.); (J.M.G.-V.)
| | - José Antonio Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD-CSIC-Universidad Pablo de Olavide), 41013 Sevilla, Spain; (P.C.-H.); (D.G.-F.); (R.P.-P.); (J.M.R.-D.); (D.R.-L.); (M.Á.-C.); (A.R.-G.); (A.L.-C.)
| |
Collapse
|
50
|
Han Q, Ni B, Bao W, Zhang J, Zheng M, Miu J, Wang Z, Yuan J, Tao J, Han Z, Gu M, Ju X, Tan R. CAV1 promotes epithelial-to-mesenchymal transition (EMT) and chronic renal allograft interstitial fibrosis by activating the ferroptosis pathway. Front Immunol 2025; 16:1523855. [PMID: 40013149 PMCID: PMC11860899 DOI: 10.3389/fimmu.2025.1523855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/21/2025] [Indexed: 02/28/2025] Open
Abstract
Background Chronic allograft dysfunction (CAD) stands as a critical factor that limits the long-term viability of transplanted kidneys. Ferroptosis is an iron-dependent form of programmed cell death increasingly linked to chronic fibrosis. However, the mechanism by which ferroptosis contributes to the onset and progression of CAD remains unclear. Methods This study analyzed transcriptome data from renal transplant biopsy samples in the Gene Expression Omnibus (GEO), through clinical samples, animal models, and cell experiments, this study investigated the mechanism by which Caveolin-1 (CAV1) promotes CAD through the regulation of the ferroptosis pathway. Results The elevated levels of CAV1 were found to positively correlate with CAD incidence. Clinical and animal model validation confirmed heightened CAV1 expression in CAD. In vitro experiments demonstrated that CAV1 can directly promote chronic renal allograft interstitial fibrosis by regulating ferroptosis in renal tubular epithelial cells; additionally, it can promote epithelial-to-mesenchymal transition (EMT) by secreting Interleukin- 6 (IL-6), thereby further contributing to CAD. Conclusion CAV1 plays a critical role in the development of CAD by promoting EMT and chronic renal allograft interstitial fibrosis through the ferroptosis pathway. Adjusting ferroptosis by altering the expression abundance of CAV1 may become an important method for the prevention and treatment of CAD in the future.
Collapse
Affiliation(s)
- Qianguang Han
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bin Ni
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Bao
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Junqi Zhang
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ming Zheng
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinxu Miu
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zijie Wang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingwen Yuan
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Tao
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhijian Han
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Gu
- Department of Urology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaobing Ju
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruoyun Tan
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|