1
|
Lu P, Zhang F, Yang L, He Y, Kong X, Guo K, Xie Y, Xie H, Xie B, Jiang Y, Peng J. Bromodomain-containing protein 4 knockdown promotes neuronal ferroptosis in a mouse model of subarachnoid hemorrhage. Neural Regen Res 2026; 21:715-729. [PMID: 39104173 DOI: 10.4103/nrr.nrr-d-24-00147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/15/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00041/figure1/v/2025-05-05T160104Z/r/image-tiff Neuronal cell death is a common outcome of multiple pathophysiological processes and a key factor in neurological dysfunction after subarachnoid hemorrhage. Neuronal ferroptosis in particular plays an important role in early brain injury. Bromodomain-containing protein 4, a member of the bromo and extraterminal domain family of proteins, participated in multiple cell death pathways, but the mechanisms by which it regulates ferroptosis remain unclear. The primary aim of this study was to investigate how bromodomain-containing protein 4 affects neuronal ferroptosis following subarachnoid hemorrhage in vivo and in vitro . Our findings revealed that endogenous bromodomain-containing protein 4 co-localized with neurons, and its expression was decreased 48 hours after subarachnoid hemorrhage of the cerebral cortex in vivo . In addition, ferroptosis-related pathways were activated in vivo and in vitro after subarachnoid hemorrhage. Targeted inhibition of bromodomain-containing protein 4 in neurons increased lipid peroxidation and intracellular ferrous iron accumulation via ferritinophagy and ultimately led to neuronal ferroptosis. Using cleavage under targets and tagmentation analysis, we found that bromodomain-containing protein 4 enrichment in the Raf-1 promoter region decreased following oxyhemoglobin stimulation in vitro . Furthermore, treating bromodomain-containing protein 4-knockdown HT-22 cell lines with GW5074, a Raf-1 inhibitor, exacerbated neuronal ferroptosis by suppressing the Raf-1/ERK1/2 signaling pathway. Moreover, targeted inhibition of neuronal bromodomain-containing protein 4 exacerbated early and long-term neurological function deficits after subarachnoid hemorrhage. Our findings suggest that bromodomain-containing protein 4 may have neuroprotective effects after subarachnoid hemorrhage, and that inhibiting ferroptosis could help treat subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Peng Lu
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Fan Zhang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Lei Yang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yijing He
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xi Kong
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Institute of Brain Science, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Kecheng Guo
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yuke Xie
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Huangfan Xie
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Institute of Brain Science, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Institute of Brain Science, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Institute of Brain Science, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| |
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
|
Yin K, Zhang X, Pei Y, Fu Y, Zhang X, Li Y, Li X, Wang X, Zhou H. Unraveling the differential tolerance mechanisms of Acropora formosa and Montipora digitata to Benzo[a]pyrene (BaP) exposure via 4D proteomics. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138213. [PMID: 40203755 DOI: 10.1016/j.jhazmat.2025.138213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 04/02/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Coral reefs are suffering from environmental pollution worldwide, implying unprecedented survival challenges. In this paper we investigate the effects of the Benzo[a]pyrene (BaP) on the survival status of coral and explore its potential tolerance mechanism. By applying advanced 4D proteomics techniques, we systematically compared the differences in the protein expression profiles of Acropora formosa (A. formosa) and Montipora digitata (M. digitata) under BaP exposure conditions (50 μg/L, 72 h and 120 h). Under the same BaP exposure conditions, the bleaching rate of A. formosa was faster, and the zooxanthellae density and chlorophyll content were lower. M. digitata showed higher BaP tolerance than A. formosa, may attributed to significantly enhanced protein synthesis, folding, and stability in its host cells, as well as a more efficient energy metabolism mechanism. While A. formosa coral hosts showed low protein stability and high ferritin expression, and iron metabolism imbalance was aggravated under BaP stress, which increased oxidative stress damage. Specifically, the zooxanthellae of M. digitata without exposed to BaP showed stronger photosynthetic efficiency and glucose metabolism, especially the activation of the pyruvate metabolic pathway. However, these advantages were rapidly diminished after exposure to BaP. In response to BaP exposure, A. formosa's zooxanthellae may activated longevity related pathways and hypoxia-inducing factor signaling pathways, significantly enhancing energy metabolism pathways. This study is helpful to reveal the complex adaptive mechanism of coral reef ecosystem to environmental pollution.
Collapse
Affiliation(s)
- Kai Yin
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Xin Zhang
- The Management Office of the National Coral Reef Nature Reserve in Sanya, Hainan 572000, China
| | - Yuebin Pei
- Cotton Research Institute,Shanxi Agriculture University, Yuncheng, Shanxi 044000, China
| | - Yijun Fu
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Xu Zhang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Yuanchao Li
- Hainan Academy of Ocean and Fisheries Sciences, Haikou 571126, China
| | - Xue Li
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Xiaobing Wang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Hailong Zhou
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China.
| |
Collapse
|
4
|
Li MH, Yang Y, Dong QQ, Tao H, Lu C, Yang JJ. Novel epitranscriptomic and epigenetic therapeutic strategies and targets for ferroptosis in liver fibrosis. Eur J Pharmacol 2025; 996:177344. [PMID: 40015597 DOI: 10.1016/j.ejphar.2025.177344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
Liver fibrosis is characterized by an excessive accumulation of extracellular matrix (ECM) and the activation of hepatic stellate cells (HSCs), which are influenced by epitranscriptomic and epigenetic factors. Recent advancements in epigenetic and epitranscriptomic research have revealed new opportunities for therapeutic interventions, particularly through the regulation of ferroptosis, a type of programmed cell death that is specifically linked to iron-dependent lipid peroxidation. In the context of liver fibrosis, a progressive scarring process that can progress to cirrhosis and ultimately end-stage liver disease, targeting these regulatory mechanisms to modulate ferroptosis presents a promising therapeutic strategy. This review aims to consolidate current knowledge on the epigenetic and epitranscriptomic control of ferroptosis and investigate its potential implications for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ming-Hui Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yang Yang
- Department of General Surgery, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
| | - Qi-Qi Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; School of Pharmacy, Anhui Medical University, Hefei, 230032, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Chao Lu
- First Affiliated Hospital, Anhui University of Science & Technology, Huainan, 232001, China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China; Center for Scientific Research and Experiment, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
5
|
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
|
6
|
Lu S, Wang P, Hu J, Zhang Z. Tetrandrine Improves Severe Acute Pancreatitis by Inhibiting NCOA4 Glycosylation-Mediated Binding to FTH1 and Inducing Autophagy-Dependent Ferroptosis. FASEB J 2025; 39:e70592. [PMID: 40326888 DOI: 10.1096/fj.202500404r] [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: 02/11/2025] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
Severe acute pancreatitis (SAP) is an acute abdominal disease with extremely high mortality; autophagy-dependent ferroptosis plays a crucial role in acute pancreatitis. However, the specific underlying mechanism remains unclear. To investigate the role of nuclear receptor coactivator 4 (NCOA4) in SAP and the mechanism by which tetrandrine influences it. Experimental SAP models were established using L-arginine (L-Arg) induction to observe changes in NCOA4 expression. Knockout and overexpression experiments of NCOA4 were conducted to assess the impact on SAP. Additionally, in vitro cell experiments were performed to verify these findings. Furthermore, the impact of N-glycosylation of NCOA4 on its function, particularly its binding ability with ferritin heavy chain 1 (FTH1), was studied. Finally, the effects of tetrandrine on N-glycosylation of NCOA4, the binding between NCOA4 and FTH1, and the progression of SAP were analyzed. NCOA4 expression was significantly upregulated in SAP. Knockout of NCOA4 improved the phenotype of SAP, whereas its overexpression exacerbated SAP. This was also confirmed in vitro. N-glycosylation of NCOA4 is crucial for its binding with FTH1, which in turn affects ferroptosis. Tetrandrine targets the N-glycosylation of NCOA4, weakening the interaction between NCOA4 and FTH1, thereby inhibiting the progression of SAP. This study demonstrates that tetrandrine targets the N-glycosylation of NCOA4, inhibiting autophagy-dependent ferroptosis mediated by its binding to FTH1 and thus ameliorates SAP. This finding provides us with a novel therapeutic approach for SAP and offers a new perspective on understanding the mechanism of action of tetrandrine in SAP.
Collapse
Affiliation(s)
- Sen Lu
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Pin Wang
- Endocrinology Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Junting Hu
- Neurosurgery Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhao Zhang
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
7
|
Liu B, Tian X, Li L, Jiang N, Cheng M, Zhu J, Wu Z. SFTSV induces liver ferroptosis through m6A-related ferritinophagy. Autophagy 2025:1-14. [PMID: 40340535 DOI: 10.1080/15548627.2025.2503564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 04/15/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is a widely prevalent infectious disease caused by severe fever with thrombocytopenia syndrome virus (SFTSV). SFTSV infection carries a high mortality rate and has emerged to be a public health concern. SFTSV infection could induce many classical cell death pathways. Ferroptosis, a novel iron-dependent form of regulated cell death, is shown to participate in various biological processes and is considered as a new therapeutic target. In the current study, we reported that SFTSV infection perturbed the classical redox cycle by downregulating the expression of GPX4, SLC7A11 and GSH, and increasing the level of reactive oxygen species (ROS) and malondialdehyde (MDA). Interestingly, we observed that the elevation of ATG5 mRNA m6A modification after SFTSV infection and mutation of the m6A-sites significantly rescued SFTSV infection-induced ferritinophagy. We further found that the NSs protein of SFTSV played a major role in driving the ferritinophagy. Finally, we found that ferroptosis inhibitor ferrostatin-1 prevented ferroptosis and suppressed SFTSV infection both in vitro and in vivo models. In summary, our study demonstrated that SFTSV infection could induce ferroptosis in liver, and m6A modified ATG5 mediated ferritinophagy to facilitate this process. Targeting ferroptosis may serve as a potential therapy for the treatment of SFTS.Abbreviations: ATG5: autophagy related 5; Baf-A1: bafilomycin A1; Fer-1: ferrostatin-1; Fe2+: ferrous iron; FTH1: ferritin heavy chain 1; GOT1/AST: glutamic-oxaloacetic transaminase 1; GPT/ALT: glutamic - pyruvic transaminase; GSH: glutathione; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MDA: malondialdehyde; NCOA4: nuclear receptor coactivator 4; ROS: reactive oxygen species; SFTSV: severe fever with thrombocytopenia virus; SQSTM1/p62: sequestosome 1.
Collapse
Affiliation(s)
- Bingxin Liu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Xiaoyan Tian
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Linrun Li
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Na Jiang
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Min Cheng
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Jin Zhu
- The Department of Immunology, Huadong Medical Institute of Biotechnique, Nanjing, China
| | - Zhiwei Wu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Q2 Science, Nanjing University, Nanjing, China
- School of Pharmacy, Dali University, Dali, Yunnan, China
| |
Collapse
|
8
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Mechanism of ferroptosis in heart failure: The role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and intervention strategies. Ageing Res Rev 2025:102770. [PMID: 40360081 DOI: 10.1016/j.arr.2025.102770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/29/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
The ferroptosis of cardiomyocytes has been recognized as the core pathological mechanism of heart failure. During the evolution of cardiovascular diseases, the accumulation of angiotensin II and advanced glycation end products can lead to the excessive activation of the RAGE/TLR4-JNK1/2 pathway, which subsequently triggers ferritinophagy, clockophagy, and enhanced p53 activity, ultimately leading to cardiomyocyte ferroptosis. It is evident that deeply unraveling the specific mechanisms in this field and comprehensively evaluating potential drugs and therapeutic strategies targeting this pathway is crucial for improving the status of cardiomyocyte ferroptosis. However, our current understanding of this pathway's specific molecular biological mechanisms in the process of cardiomyocyte ferroptosis remains limited. In light of this, this paper first comprehensively reviews the historical context of ferroptosis research, compares the similarities and differences between ferroptosis and other standard modes of cell death, elucidates the core mechanisms of ferroptosis and its close connection with heart failure, aiming to establish a basic cognitive framework for readers on ferroptosis and its role in heart failure. Subsequently, the paper delves into the pivotal role of the RAGE/TLR4-JNK1/2 pathway in cardiomyocyte ferroptosis and its intricate molecular biological regulatory network. Furthermore, it systematically integrates various therapeutic approaches aimed at inhibiting RAGE, TLR4, and JNK1/2 activity to alleviate cardiomyocyte ferroptosis, encompassing RNA interference technology, gene knockout techniques, small molecule inhibitors, natural active ingredients, as well as traditional Chinese and Western medicines, with the ultimate goal of forging new avenues and strategies for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, PR China.
| |
Collapse
|
9
|
Liu RQ, Wu YT, Cheng Y, Chang YH, Saleem MAU, Hu ZY, Yang SJ, Wang XQ, Song YJ, Mao XY, Zheng J, Wang YB, Lou M, Zhao Y, Li JL. TBBPA induced hepatocyte ferroptosis by PCBP1-mediated ferritinophagy. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138515. [PMID: 40359755 DOI: 10.1016/j.jhazmat.2025.138515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/26/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025]
Abstract
Tetrabromobisphenol A (TBBPA) is the most widely used brominated flame retardant and has been identified as emerging widespread pollutants. Ferroptosis, a recently characterized form of iron-dependent cell death, is related to a wide range of liver diseases. Ferritinophagy as a novel selective form of autophagy functions in iron processing is essential to induce ferroptosis. Poly(rC)-binding protein 1 (PCBP1) is an iron chaperone involved in iron loading to ferritin. Nevertheless, the potential health risk caused by TBBPA in mammals is unknown. Thus, this study is conducted to explore the molecular mechanism of TBBPA-induced liver injury and the unique role of PCBP1 in it. In this study, we found that TBBPA exposure caused hepatic pathological injury and hepatocyte mitochondrial morphological changes, such as decreased or absent mitochondrial crest, ruptured mitochondrial membranes and mitochondrial shrinkage. The result showed that TBBPA exposure exacerbated glutathione depletion and lipid peroxidation, which are hallmarks of ferroptosis. Consistent with the results in vivo, TBBPA exposure activated ferritinophagy and upregulated indicators related to ferroptosis in hepatocytes. Of note, overexpression of PCBP1 inhibited TBBPA-induced ferroptosis by reducing overstimulated ferritinophagy. Here, we uncover a new mechanism whereby TBBPA triggers hepatocyte ferroptosis through the activation of ferritinophagy. Of note, we identify PCBP1 as critical for liver iron homeostasis, link this molecule to liver disease. Taken together, our findings provide a new therapeutic strategy and potential target for the treatment of liver disease.
Collapse
Affiliation(s)
- Rui-Qi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yu-Tong Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yue Cheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yuan-Hang Chang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | | | - Zi-Yan Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shang-Jia Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xue-Qi Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yi-Jia Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xin-Yue Mao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jing Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yi-Bo Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Ming Lou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yi Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China
| |
Collapse
|
10
|
Chandrasekaran K, Lee CE, Yun S, Jangid AK, Kim S, Kim K. CD44 Receptor-Mediated Ferroptosis Induction by Hyaluronic Acid Carbon Quantum Dots in Triple-Negative Breast Cancer Cells Through Downregulation of SLC7A11 Pathway. MATERIALS (BASEL, SWITZERLAND) 2025; 18:2139. [PMID: 40363642 DOI: 10.3390/ma18092139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
The field of cancer therapy is actively pursuing highly effective self-targeted drug delivery materials endowed with exceptional properties. Recently, hyaluronic acid (HA), a naturally occurring polysaccharide, has been recognized as a potential target ligand for CD44 receptors, which are frequently expressed on various solid tumor cells targeted in cancer therapy. HA carbon quantum dots (CQDs) exhibit several advantageous properties, including a high surface area-to-volume ratio, small particle size, biocompatibility, and low cytotoxicity, making them ideal for biomedical applications, such as CD44-targeted drug delivery in ferroptosis-based cancer therapy. In this study, we synthesized HA-CQDs to enhance CD44-mediated ligand-receptor interactions targeting triple-negative breast cancer (TNBC). CQDs facilitate the intracellular generation of reactive oxygen species (ROS), leading to glutathione depletion. These processes result in crucial actions such as the downregulation of glutathione peroxidase 4, downregulation of solute carrier family 7 member 11, and inhibition of cystine intake. The subsequent intracellular ROS, originating from lipid peroxidation, induces ferroptosis. Our HA-CQDs engage CD44 receptors, selectively targeting TNBCs and enhancing cancer recognition. This interaction potentially enhances the nanoplatform-based CD44 targeted therapeutic effects in inducing ferroptosis.
Collapse
Affiliation(s)
| | - Chae Eun Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Seojeong Yun
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
11
|
Liang SM, Shen J, Ma RT, Du ND, Wang R, Wu ZM, Shan M, Liang SR, Hu WR, Wang W, Sheng WW, Huang DF, Chen XH. Ferritinophagy-derived iron causes protein nitration and mitochondrial dysfunction in acetaminophen-induced liver injury. Toxicol Appl Pharmacol 2025; 500:117376. [PMID: 40339610 DOI: 10.1016/j.taap.2025.117376] [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: 01/07/2025] [Revised: 04/23/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Acetaminophen (APAP), also known as paracetamol, is a widely used analgesic and antipyretic drug. While the drug is effective and safe at recommended doses, excessive intake can lead to acute liver injury (ALI) due to the formation of the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI), which depletes glutathione (GSH). Despite regulatory efforts, APAP-related liver injury remains a significant health concern. However, the cellular pathways that contribute to APAP-induced hepatotoxicity-particularly those involving iron metabolism-remain incompletely understood. To address this gap, we investigated whether ferritinophagy-the autophagic degradation of ferritin heavy chain (FTH) mediated by nuclear receptor coactivator 4 (NCOA4)-contributes to APAP-induced ALI. We administered APAP to C57BL/6 J mice and AML-12 hepatocyte cells and monitored markers of ferritinophagy, iron release, and hepatic injury. In parallel, we assessed the protective effect of the iron chelator deferoxamine (DFO) to validate the pathogenic role of free iron in vivo. First, in vivo studies revealed that APAP treatment significantly upregulated NCOA4 and FTH mRNA expression at 6 h post-exposure, coupled with increased LC3II protein and decreased p62, NCOA4, and FTH protein levels-hallmarks of active ferritinophagy. Importantly, pretreatment of mice with DFO markedly attenuated serum ALT elevation and histopathological liver damage, indicating that iron released via ferritinophagy critically mediates APAP-induced hepatotoxicity. To corroborate these findings at the cellular level, we measured free iron and ferritinophagy-related proteins in AML-12 cells following APAP exposure. We observed a progressive increase in free iron, with FTH protein level peaking at 2 h and subsequently declining by 6 and 12 h. Concurrently, LC3II protein level rose while NCOA4 protein decreased at 6 h, confirming activation of ferritinophagy in vitro. Although canonical ferroptosis is driven by iron-catalyzed lipid peroxidation (LPO), our APAP model did not exhibit key ferroptotic signatures. In vivo, malondialdehyde (MDA) level and Ptgs2 mRNA did not increase significantly, nor did GPX4 protein level decrease after APAP administration. Similarly, AML-12 cells failed to show a significant rise in C11-BODIPY oxidation after APAP treatment. Thus, APAP-induced ferritinophagy doesn't result in significant LPO. Instead of LPO, APAP exposure led to pronounced protein nitration and mitochondrial dysfunction. Specifically, the protein level of nitrotyrosine (NT) increased significantly at 6 h in vivo, while AML-12 cells exhibited elevated mitochondrial reactive oxygen species (MtROS) alongside reduced mitochondrial membrane potential (MMP) and ATP level. Collectively, these data suggest that ferritinophagy-derived iron triggers protein nitration and mitochondrial impairment, culminating in cell death. Given NCOA4's central role in ferritinophagy, we next evaluated whether its knock-down could mitigate APAP-induced mitochondrial dysfunction. NCOA4 siRNA in AML-12 cells restored ATP level, enhanced MMP, and reduced Fe2+ accumulation and MtROS generation after APAP treatment. Overall, our findings illuminate ferritinophagy-derived iron as a critical driver of APAP hepatotoxicity and nominate NCOA4 inhibition as a promising therapeutic strategy against APAP-induced ALI.
Collapse
Affiliation(s)
- Shi-Min Liang
- Department of Gastroenterology, Luohe Central Hospital, Luohe 462000, Henan Province, China
| | - Jie Shen
- Department of Gastroenterology, Luohe Central Hospital, Luohe 462000, Henan Province, China; Fujian Medical University, Fuzhou 350000, Fujian Province, China
| | - Rui-Ting Ma
- Department of Gastroenterology, Luohe Central Hospital, Luohe 462000, Henan Province, China; Henan University, Zhengzhou 450000, Henan Province, China
| | - Nan-Di Du
- Traditional Chinese Medicine Preparation Modern Technology Research and Development and Clinical Application Engineering Center of Henan Province, Luohe 462000, Henan Province, China; Key Laboratory of Traditional Chinese Medicine Preparation and Processing of Henan Province, Luohe 462000, Henan Province, China
| | - Rui Wang
- Traditional Chinese Medicine Preparation Modern Technology Research and Development and Clinical Application Engineering Center of Henan Province, Luohe 462000, Henan Province, China; Key Laboratory of Traditional Chinese Medicine Preparation and Processing of Henan Province, Luohe 462000, Henan Province, China
| | - Zuo-Min Wu
- Traditional Chinese Medicine Preparation Modern Technology Research and Development and Clinical Application Engineering Center of Henan Province, Luohe 462000, Henan Province, China; Key Laboratory of Traditional Chinese Medicine Preparation and Processing of Henan Province, Luohe 462000, Henan Province, China
| | - Min Shan
- Department of Neurology, Luohe Central Hospital, Luohe 462000, Henan Province, China
| | - Shi-Rong Liang
- Nanyang Institute of Technology, Nanyang 473000, Henan Province, China
| | - Wei-Rong Hu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei Wang
- Department of Obstetrics and Gynaecology, Luohe Central Hospital, Luohe 462000, Henan, China; Henan Key Laboratory of Fertility Protection and Aristogenesis, Luohe 462000, Henan, China
| | - Wei-Wei Sheng
- Nanjing Drum Tower Hospital, Nanjing 210000, Jiangsu Province, China
| | - De-Feng Huang
- Department of Gastroenterology, Luohe Central Hospital, Luohe 462000, Henan Province, China.
| | - Xiao-Hua Chen
- Department of Gastroenterology, Luohe Central Hospital, Luohe 462000, Henan Province, China.
| |
Collapse
|
12
|
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
|
13
|
Wang X, Liu Z, Lin C. Metal ions-induced programmed cell death: how does oxidative stress regulate cell death? Life Sci 2025; 374:123688. [PMID: 40328371 DOI: 10.1016/j.lfs.2025.123688] [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: 11/17/2024] [Revised: 04/20/2025] [Accepted: 05/01/2025] [Indexed: 05/08/2025]
Abstract
In recent years, the mechanisms of ferroptosis and cuproptosis, two novel modes of cell death, have been elucidated and have attracted much attention. Ferroptosis is dependent on the metabolic disruption of iron ions and lipid peroxidation, whereas cuproptosis is closely related to intracellular accumulation of copper ions, aggregation of lipoylated proteins and damage to FeS cluster proteins. In particular, oxidative stress plays an important role in both types of cell death. During ferroptosis, the central role of oxidative stress is reflected in the overproduction of reactive oxygen species (ROS) and lipid peroxidation of the cell membrane. Recent studies have revealed that ROS can propagate over long distances across cells in the form of trigger waves, triggering large-scale ferroptosis. In embryonic development, different regional redox states can limit the long-distance propagation of ferroptosis waves, which is critical for muscle remodeling and tissue formation during development. In cuproptosis, processes such as copper ions accumulation, tricarboxylic acid (TCA) cycle blockade, and reduced level of FeS cluster proteins are closely associated with oxidative stress. In addition, there is a close link between oxidative stress and death induced by other metal ions (Ca2+, Zn2+, etc.). In this paper, we review the role of oxidative stress in ferroptosis and cuproptosis and the related research progress to provide new ideas for understanding the mechanism of cell death and the occurrence and treatment of related diseases.
Collapse
Affiliation(s)
- Xingsheng Wang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Zuohao Liu
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Changjun Lin
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
14
|
Di Lorenzo R, Marzetti E, Coelho-Junior HJ, Calvani R, Pesce V, Landi F, Leeuwenburgh C, Picca A. Iron Metabolism and Muscle Aging: Where Ferritinophagy Meets Mitochondrial Quality Control. Cells 2025; 14:672. [PMID: 40358196 DOI: 10.3390/cells14090672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
In older adults with reduced physical performance, an increase in the labile iron pool within skeletal muscle is observed. This accumulation is associated with an altered expression of mitochondrial quality control (MQC) markers and increased mitochondrial DNA damage, supporting the hypothesis that impaired MQC contributes to muscle dysfunction during aging. The autophagy-lysosome system plays a critical role in MQC by tagging and engulfing proteins and organelles for degradation in lysosomes. The endolysosomal system is also instrumental in transferrin recycling, which, in turn, regulates cellular iron uptake. In the neuromuscular system, the autophagy-lysosome system supports the structural integrity of neuromuscular junctions, and its dysfunction contributes to muscle atrophy. While MQC was thought to protect against iron-induced cell death, the discovery of ferroptosis, a form of iron-dependent cell death, has highlighted a complex interplay between MQC and iron-inflicted damage. Ferritinophagy, the autophagic degradation of ferritin, if overactivated, can induce ferroptosis. Alternatively, aging may impair ferritinophagy, leading to ferritin accumulation and the release of toxic labile iron under stress, exacerbating oxidative damage and cellular senescence. Physical activity supports muscle health also by preserving mitochondrial quantity and quality and enhancing bioenergetics. However, therapeutic strategies for preventing or reversing physical function decline in aging are still lacking due to the insufficient understanding of the underlying mechanisms. Unveiling how disruptions in iron homeostasis impact muscle quality in older adults may allow for the development of therapeutic strategies targeting iron handling to alleviate age-associated muscle decline.
Collapse
Affiliation(s)
- Rosa Di Lorenzo
- Department of Biosciences, Biotechnologies, and Environment, Università degli Studi di Bari Aldo Moro, Via Edoardo Orabona 4, 70125 Bari, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Helio José Coelho-Junior
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Vito Pesce
- Department of Biosciences, Biotechnologies, and Environment, Università degli Studi di Bari Aldo Moro, Via Edoardo Orabona 4, 70125 Bari, Italy
| | - Francesco Landi
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Christiaan Leeuwenburgh
- Department of Physiology and Aging, University of Florida, 2004 Mowry Road, Gainesville, FL 32611, USA
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Department of Medicine and Surgery, LUM University, Str. Statale 100, 70010 Casamassima, Italy
| |
Collapse
|
15
|
Fan C, Luo Z, Zheng Q, Xu Y, Xu Y, Chen J, Meng Y, Jiang H, Liu K, Xi Y. Cytoglobin augments ferroptosis through autophagic degradation of ferritin in colorectal cancer cells. Mol Cell Biochem 2025; 480:2881-2892. [PMID: 39503803 PMCID: PMC12048458 DOI: 10.1007/s11010-024-05148-0] [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: 09/21/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024]
Abstract
Autophagy has gained importance in the context of ferroptosis. Nevertheless, a deeper understanding of the regulatory mechanism governing autophagy-dependent ferroptosis is necessary. Cytoglobin (CYGB), a member of the globin family, exhibits antifibrotic effects, regulates cellular reactive oxygen species, and stimulates tumor inhibition. Herein, we present further insights into the role of CYGB in ferroptosis regulation. Our investigation confirmed that CYGB impedes cell proliferation and migration. Furthermore, a significant association between CYGB and the lysosomal pathway was suggested based on the RNA sequencing data analysis. Elevated lysosomal signal and colocalization of CYGB with lysosome-associated membrane glycoprotein 1 (LAMP1) were observed. Moreover, upregulated autophagy and augmented ferroptosis induced by RSL3 were confirmed in CYGB-overexpression cells with an obviously increased colocalization of nuclear receptor coactivator 4 (NCOA4) and LC3B. The autophagy inhibitor bafilomycin or chloroquine alleviated autophagy-dependent degradation of ferritin protein under RSL3 treated condition. Additionally, a colocalization of CYGB with the transferrin receptor (TFR) was confirmed. Our results demonstrate an important functional pathway by which CYGB regulates ferroptosis through TFR-binding and autophagic degradation of ferritin, and provide a potential pathway for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Chengjiang Fan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Ziyang Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
- Institute of Transplantation Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Qingfang Zheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yuhang Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yao Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jianing Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - You Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Haizhong Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, China
| | - Kaitai Liu
- Department of Radiation Oncology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315001, China
| | - Yang Xi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
16
|
Seke M, Stankovic A, Zivkovic M. Capacity of fullerenols to modulate neurodegeneration induced by ferroptosis: Focus on multiple sclerosis. Mult Scler Relat Disord 2025; 97:106378. [PMID: 40088719 DOI: 10.1016/j.msard.2025.106378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/10/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
Multiple sclerosis is an inflammatory disease of the central nervous system (CNS), characterized by oligodendrocyte loss and demyelination of axons leading to neurodegeneration and severe neurological disability. Despite the existing drugs that have immunomodulatory effects an adequate therapy that slow down or stop neuronal death has not yet been found. Oxidative stress accompanied by excessive release of iron into the extracellular space, mitochondrial damage and lipid peroxidation are important factors in the controlled cell death named ferroptosis, latterly recognized in MS. As the fullerenols exhibit potent antioxidant activity, recent results imply that they could have protective effects by suppressing ferroptosis. Based on the current knowledge we addressed the main mechanisms of the protective effects of fullerenols in the CNS in relation to ferroptosis. Inhibition of inflammation, iron overload and lipid peroxidation through the signal transduction mechanism of Nuclear Factor Erythroid 2-Related Factor 2 (NRF2), chelation of heavy metals and free radical scavenging using fullerenols are proposed as benefitial strategy preventing MS progression. Current review connects ferroptosis molecular targets and important factors of MS progression, with biomedical properties and mechanisms of fullerenols' actions, to propose new treatment strategies that could be addaptobale in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Mariana Seke
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia.
| |
Collapse
|
17
|
Jian PA, Yang TN, Wang YX, Ma XY, Huang NN, Ren YF, Yuan SH, Li JL, Wang CC, Li XN. Lycopene, a natural plant extract, alleviates atrazine-induced ferroptosis in hepatocytes by activating cytochrome P450 oxidoreductase. Int J Biol Macromol 2025; 308:142311. [PMID: 40139611 DOI: 10.1016/j.ijbiomac.2025.142311] [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] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Atrazine (ATZ) and diaminochlorotriazine (DACT) accumulation poses liver health risks in animals and humans. Lycopene (LYC), a carotenoid found in red plants and fruits, exhibits potent antioxidant effects. This study explores the interaction between LYC and ATZ in mouse hepatocyte ferroptosis and the potential regulatory role of Cytochrome P450 oxidoreductase (CYPOR) in this process. Male mice were exposed to ATZ (50 mg/kg or 200 mg/kg) and/or LYC (5 mg/kg) by gavage for 21 days. In vitro experiments, a mouse hepatocyte cell line (AML12) was exposed to DACT (200 μM) and/or LYC (2 μM) for 12 h with or without small interfering RNA treatment. We found that both ATZ and DACT promoted CYPOR expression and caused liver injury. ATZ/DACT promotes Fe2+ accumulation and lipid peroxidation, ultimately leading to Ferroptosis in mouse hepatocytes. However, LYC alleviated ATZ/DACT-induced Ferroptosis by inhibiting CYPOR. The CYPOR knockdown resulted in the blockage of ATZ/DACT-induced ferroptosis, while the alleviation of ferroptosis by LYC was further enhanced. Thus, CYPOR can regulate ferroptosis in mouse hepatocytes and is a novel target for the treatment of hepatocyte ferroptosis-related diseases. Lycopene can be used as a functional dietary supplement to scavenge ferroptosis and reduce chronic liver disease.
Collapse
Affiliation(s)
- Ping-An Jian
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Tian-Ning Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yu-Xiang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiang-Yu Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Ning-Ning Huang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yi-Fei Ren
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shi-Hao Yuan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China
| | - Chi-Chiu Wang
- Department of Obstetrics & Gynaecology, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, The Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong.
| | - Xue-Nan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Department of Obstetrics & Gynaecology, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, The Chinese University of Hong Kong-Sichuan University Joint Laboratory for Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
18
|
Nie T, Fang Y, Zhang R, Cai Y, Wang X, Jiao Y, Wu J. Self-healable and pH-responsive spermidine/ferrous ion complexed hydrogel Co-loaded with CA inhibitor and glucose oxidase for combined cancer immunotherapy through triple ferroptosis mechanism. Bioact Mater 2025; 47:51-63. [PMID: 39877156 PMCID: PMC11772096 DOI: 10.1016/j.bioactmat.2025.01.005] [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: 07/01/2024] [Revised: 12/24/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Tumor microenvironment governs various therapeutic tolerability of cancer such as ferroptosis and immunotherapy through rewiring tumor metabolic reprogramming like Warburg metabolism. Highly expressed carbonic anhydrases (CA) in tumor that maintaining the delicate metabolic homeostasis is thus the most potential target to be modulated to resolve the therapeutic tolerability. Hence, in this article, a self-healable and pH-responsive spermidine/ferrous ion hydrogel loaded with CA inhibitor (acetazolamide, ACZ) and glucose oxidase (ACZ/GOx@SPM-HA Gel) was fabricated through the Schiff-base reaction between spermidine-dextran and oxidized hyaluronic acid, along with ferrous coordination. Investigation on cancer cell lines (MOC-1) demonstrated ACZ/GOx@SPM-HA Gel may induce cellular oxidative stress and mitochondrial dysfunction through disrupting the cellular homeostasis. Moreover, with the facilitation of autophagy induced by spermidine, ACZ/GOx@SPM-HA Gel may trigger a positive feedback loop to maximally amplify cellular ferroptosis and promote DAMPs release. The anti-tumor evaluation on xenograft mice models furtherly proved the local injection of such hydrogel formulation could efficiently inhibit the tumor growth and distinctively promote the immunogenicity of tumor bed to provide a more favorable environment for immunotherapy. Overall, ACZ/GOx@SPM-HA Gel, with such feasible physiochemical properties and great biocompatibility, holds great potential in treating solid tumors with acidosis-mediated immunotherapy tolerance.
Collapse
Affiliation(s)
- Tianqi Nie
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
| | - Yifei Fang
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ruhe Zhang
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518106, China
| | - Yishui Cai
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, 511400, China
| | - Xiaobo Wang
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518106, China
| | - Yuenong Jiao
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou Twelfth People's Hospital (The Affiliated Twelfth People's Hospital of Guangzhou Medical University), Guangzhou Medical University, Guangzhou, 510620, China
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou, 511400, China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
- Department of Hematology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518106, China
| |
Collapse
|
19
|
Shyr ZA, Amniouel S, Owusu-Ansah K, Tambe M, Abbott J, Might M, Zheng W. Increased oxidative stress and autophagy in NGLY1 patient iPSC-derived neural stem cells. Exp Cell Res 2025; 448:114540. [PMID: 40189184 PMCID: PMC12070446 DOI: 10.1016/j.yexcr.2025.114540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
NGLY1 (N-glycanase) is a de-glycosylating enzyme that promotes clearance of misfolded glycan proteins. NGLY1 deficiency leads to a disease pathology with varied symptoms, including severe neurological defects. There are no therapeutic options currently available for the treatment of this rare disease. With the goal of finding potential therapeutic avenues, we performed comprehensive characterization of aberrant cellular stress pathways in a patient relevant model of NGLY1 deficiency. For a more accurate study of NGLY1 deficiency without other confounding factors, we compared differences between iPSC-derived neural stem cells carrying the commonly occurring nonsense mutation c.1201A > T (p.R401X) and their genetically similar CRISPR-corrected isogenic controls. Our findings demonstrate that NGLY1 deficiency in neural stem cells leads to an upregulation of ER stress, increased autophagic flux and significant signs of oxidative stress. These results provide new insights into the cellular dysfunctions associated with this disorder. Moreover, they point to better establishing reliable high throughput phenotypic assays that can be utilized for drug discovery.
Collapse
Affiliation(s)
- Zeenat A Shyr
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| | - Soukaina Amniouel
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Kofi Owusu-Ansah
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Mitali Tambe
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Joshua Abbott
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Matthew Might
- Hugh Kaul Precision Medicine Institute, The University of Alabama at Birmingham, AL, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
20
|
Wang P, Niu T, Huang D, Li Y, Jiang Z, Wang X, Liao L. Molecular mechanism of programmed cell death in drug-induced neuronal damage: A special focus on ketamine-induced neurotoxicity. Toxicology 2025; 513:154102. [PMID: 40015548 DOI: 10.1016/j.tox.2025.154102] [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: 01/01/2025] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
In recent years, the abuse of ketamine as a recreational drug has been growing, and has become one of the most widely abused drugs. Continuous using ketamine poses a risk of drug addiction and complications such as attention deficit disorder, memory loss and cognitive decline. Ketamine-induced neurotoxicity is thought to play a key role in the development of these neurological complications. In this paper, we focus on the molecular mechanisms of ketamine-induced neurotoxicity. According to our analyses, drugs in causing neurotoxicity are closely associated with programmed cell death (PCD) such as apoptosis, autophagy, necroptosis, pyroptosis, and Ferroptosis. Therefore, this review will collate the existing mechanisms of programmed death in ketamine-induced neurotoxicity as well as explore the possible mechanisms by outlining the mechanisms of programmed death in other drug-induced neurotoxicity, which may be helpful in identifying potential therapeutic targets for neurotoxicity induced by ketamine abuse.
Collapse
Affiliation(s)
- Peipei Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Tong Niu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Degao Huang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yuanlong Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zihan Jiang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Xia Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| | - Linchuan Liao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
21
|
Guo S, Zhang Q, Ge H, Wang H. Baicalin plays a protective role by regulating ferroptosis in multiple diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4837-4849. [PMID: 39661143 DOI: 10.1007/s00210-024-03704-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
Ferroptosis is a new kind of cell death discovered in recent years, usually accompanied by a large number of lipid peroxidation and iron accumulation in the process of cell death. Ferroptosis has been proven to play an important role in various diseases, including ischemic reperfusion injury, cancer, and neurodegeneration. Therefore, the regulation of ferroptosis will have a vital impact on the occurrence and development of diseases. Baicalin is a flavonoid compound extracted and isolated from the dried roots of Scutellaria baicalensis Georgi, a plant in the family Lamiaceae. It has various biological activities such as antioxidant, anti-proliferative, anti-inflammatory, anti-thrombotic, and regulates apoptosis and ferroptosis. Recently, increasing evidence indicates that baicalin regulation of ferroptosis is involved in multiple diseases. However, the relevant mechanisms are not yet fully understood. Here, we summarized the role of baicalin regulation of ferroptosis in different kinds of diseases, and conducted an in-depth analysis of the relevant mechanisms, hoping to provide the theoretical references for future related researches.
Collapse
Affiliation(s)
- Shiyun Guo
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Qi Zhang
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Hangwei Ge
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China
| | - Honggang Wang
- Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
22
|
Suchiita A, Gupta N, Nandi K, Goswami B. Navigating the crossroads of cell death interplay and intersections among ferroptosis, apoptosis and autophagy. Drug Metab Pers Ther 2025:dmdi-2024-0073. [PMID: 40278507 DOI: 10.1515/dmpt-2024-0073] [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: 09/14/2024] [Accepted: 01/28/2025] [Indexed: 04/26/2025]
Abstract
The review article, "Navigating the Crossroads of Cell Death: Interplay and Intersections Among Ferroptosis, Apoptosis, and Autophagy," delves into the complex interactions between these three key cell death pathways. Understanding how ferroptosis, apoptosis, and autophagy intersect is crucial for maintaining cellular homeostasis. Each pathway represents a unique mechanism of cell death, and recent research highlights their intricate interconnections and mutual influences. Exploring these relationships is vital for comprehending how cells make fate decisions and how these processes are implicated in various diseases. The review's significance lies in elucidating the molecular details of cell death and providing insight into how cells balance survival and death. The interplay among ferroptosis, apoptosis, and autophagy has important implications for developing therapeutic interventions, particularly in diseases where cell death regulation is disrupted. By examining the molecular crosstalk between these pathways, researchers can identify new drug targets and devise strategies to modulate cell fate effectively. This review aims to enhance our understanding of cell biology by offering a detailed perspective on the dynamic and interconnected nature of these cell death mechanisms.
Collapse
Affiliation(s)
| | - Navya Gupta
- 28862 Maulana Azad Medical College , New Delhi, India
| | - Kajal Nandi
- 28862 Maulana Azad Medical College , New Delhi, India
| | | |
Collapse
|
23
|
Wu C, Deng K, Zhang Y, Qin Y, Wen J, Chen BT, Jiang M. Advanced neuroimaging in systemic lupus erythematosus: identifying biomarkers for cognitive dysfunction. Neuroradiology 2025:10.1007/s00234-025-03619-9. [PMID: 40293471 DOI: 10.1007/s00234-025-03619-9] [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: 01/07/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Cognitive dysfunction (CD) is a common manifestation of central nervous system involvement in patients with systemic lupus erythematosus (SLE). Patients with SLE may develop CD insidiously at an early stage of the disease, and the lack of a standardized diagnostic test poses a major challenge in prompt diagnosis and management of these patients. This review summaries the current application of various magnetic resonance imaging (MRI) techniques for patients with SLE complicated with CD, aiming to identify potential quantitative neuroimaging biomarkers for patients with SLE and CD. METHODS We systematically searched several databases between January 2003 to December 2024. We screened retrospective and prospective studies based on search criteria keywords (including structural or functional MRI, cognitive function, lupus, and systemic lupus erythematosus) to identify peer-reviewed articles that reported advanced structural and functional MRI metrics and evaluated CD in human patients with SLE. RESULTS 123 studies (19 Bold-MRI studies, 9 DTI studies, 2 ASL studies, 4 MTI studies, 5 machine learning, and 84 other studies) were identified. Neuroimaging findings show that patients with CD have abnormal manifestations in the limbic system, hippocampus, corpus callosum, and frontal cortex, and these manifestations are closely related to cognitive functions. The most commonly affected cognitive domains are memory, attention, and executive ability. Multimodal MRI, integrating structural, functional, and perfusion parameters, combined with machine learning, can effectively predict cognitive function. CONCLUSION Advanced MRI analysis can identify the abnormalities in the whole brain and local brain regions associated with CD in patients with SLE. The integration of machine learning and multimodal MRI offers new perspectives for early identification and mechanistic studies of CD in SLE patients. More studies are needed to identify potential neuroimaging biomarkers to facilitate early diagnosis, timely treatment, and accurate prognosis for SLE patients with CD.
Collapse
Affiliation(s)
- Chengli Wu
- First Affiliated Hospital of GuangXi Medical University, Nanning, China
| | - Kemei Deng
- First Affiliated Hospital of GuangXi Medical University, Nanning, China
| | - Yu Zhang
- First Affiliated Hospital of GuangXi Medical University, Nanning, China
| | - Yuhong Qin
- First Affiliated Hospital of GuangXi Medical University, Nanning, China
| | - Jing Wen
- First Affiliated Hospital of GuangXi Medical University, Nanning, China
| | - Bihong T Chen
- City of Hope National Medical Center, Duarte, CA, USA
| | - Muliang Jiang
- First Affiliated Hospital of GuangXi Medical University, Nanning, China.
| |
Collapse
|
24
|
Liu W, Fu D, Di C, Sun J, Liu P. Autophagy is involved in Salmonella Typhimurium-induced ferroptosis in macrophages. Vet Microbiol 2025; 305:110538. [PMID: 40311251 DOI: 10.1016/j.vetmic.2025.110538] [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: 05/29/2024] [Revised: 04/21/2025] [Accepted: 04/26/2025] [Indexed: 05/03/2025]
Abstract
Salmonella is one of the most common zoonotic pathogens, posing a significant threat to both animal and human health. Our previous study demonstrated that autophagy plays a crucial role in restricting the intracellular growth of Salmonella. This study aims to investigate the effect of autophagy in Salmonella Typhimurium (S. Typhimurium)-induced ferroptosis. First, we found that S. Typhimurium induced lipid peroxidation by increasing intracellular Fe2 + levels, promoting lipid oxidation, and inhibiting the antioxidant pathway. S. Typhimurium-induced lipid peroxidation led to ferroptosis in macrophages. Further results revealed that S. Typhimurium triggered ferritin degradation by NCOA4-mediated ferritinophagy. Additionally, S. Typhimurium-induced chaperone-mediated autophagy (CMA) degraded GPX4 through TAK1-HSC70 signaling pathway. Notably, GPX4 is involved in intracellular S. Typhimurium release. Overall, autophagy was essential for S. Typhimurium induced-ferroptosis, TAK1 not only facilitated autophagy to eliminate intracellular bacteria but also promoted bacterial release.
Collapse
Affiliation(s)
- Wei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu Province 225009, China.
| | - Daobin Fu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| | - Chuanyuan Di
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| | - Jing Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| | - Penggang Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu Province 225009, PR China
| |
Collapse
|
25
|
Ke A, Yang W, Zhang W, Chen Y, Meng X, Liu J, Dai D. The cardiac glycoside periplocymarin sensitizes gastric cancer to ferroptosis via the ATP1A1-Src-YAP/TAZ-TFRC axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156804. [PMID: 40311597 DOI: 10.1016/j.phymed.2025.156804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Targeting ferroptosis vulnerabilities in tumors has become an increasingly promising therapeutic strategy. While the regulatory effects of natural products on ferroptosis are progressively being elucidated, the role of cardiac glycosides in modulating ferroptosis remains poorly understood. PURPOSE This study aims to investigate the ferroptosis-sensitizing effects of periplocymarin (PPM), a cardiac glycoside derived from the traditional plant Periploca sepium, and to elucidate the underlying molecular mechanisms. METHODS The effects of PPM on ferroptosis regulation were comprehensively assessed through functional assays, followed by sequencing analysis to identify associated signaling pathways. Subsequent mechanistic validation experiments were conducted to confirm the upstream and downstream regulatory components involved in this ferroptosis-modulating axis. RESULTS PPM induced slow and mild apoptosis in gastric cancer cells through the inhibition of glycolysis. However, when combined with ferroptosis inducers, it promoted rapid and robust ferroptosis. In vivo, PPM sensitized gastric cancer xenografts to cisplatin-induced ferroptosis with no observable cardiotoxicity or renal impairment. Mechanistically, PPM targeted the α1 subunit of the Na+/K+-ATPase (ATP1A1), leading to the activation of Src, which subsequently induced tyrosine phosphorylation of YAP/TAZ in a Hippo-independent manner, promoting their nuclear translocation. The YAP/TAZ-TEAD transcriptional complex directly bound to the TFRC promoter region between nucleotides 401-409 upstream of the transcription start site, thereby activating TFRC transcription. This resulted in increased iron influx, elevated lipid peroxidation, and heightened sensitivity to ferroptosis. Notably, ATP1A1 was essential for ferroptosis resistance, as its knockdown mimicked the sensitizing effect of PPM on ferroptosis. Moreover, the oncogenic Src-YAP/TAZ-TFRC axis may have represented a ferroptosis vulnerability and a potential biomarker in ferroptosis therapy for cancer. Importantly, other cardiac glycosides targeting Na+/K+-ATPase, such as digitoxin and bufalin, also enhanced ferroptosis sensitivity in gastric cancer cells through activation of YAP/TAZ signaling. CONCLUSION Our findings establish the cardiac glycoside PPM as a novel ferroptosis sensitizer that targets ATP1A1 to activate the Src-YAP/TAZ-TFRC axis, providing mechanistic insights for repurposing cardiac glycosides as ferroptosis modulators in precision combinatorial cancer therapy.
Collapse
Affiliation(s)
- Angting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Weiguang Yang
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wanchuan Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yibin Chen
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xiangyu Meng
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Jie Liu
- Translational Research Experiment Department, Science Experiment Center, China Medical University, Shenyang 110122, China
| | - Dongqiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
26
|
Wu Y, Yin L, Wang Z, Yuan S, Ma D, Wen C, Tian H, Xiao B, Ma C, Song L. Hydroxysafflor yellow A inhibits neuronal ferroptosis and ferritinophagy in ischemic stroke. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167867. [PMID: 40280202 DOI: 10.1016/j.bbadis.2025.167867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 02/26/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Ischemic stroke is a significant cause of disability and mortality on a global scale, with neuronal dysfunction playing a critical role in its pathogenesis. Conventional treatment approaches for ischemic stroke involve surgical interventions and thrombolytic therapy, yet these methods frequently result in ischemia/reperfusion (I/R) injury. Recent studies have underscored the implication of diverse programmed cell death mechanisms, including ferroptosis, in the progression of ischemic stroke. Ferroptosis, a newly recognized form of cell death reliant on iron, is intricately linked to various neurological conditions. Despite the existing body of research on ferritinophagy and neuronal ferroptosis in the context of cerebral ischemia-reperfusion injury, there is a lack of understanding regarding the mechanisms involved in neuronal ferroptosis. This study seeks to explore the relationship between neuronal autophagy and neuronal ferroptosis using in vivo and in vitro models of cerebral ischemia/reperfusion. The findings of our study reveal a significant upregulation of the ferritinophagy-associated protein NCOA4 following cerebral ischemia/reperfusion, concomitant with the initiation of ferroptosis in neuronal cells. This observation offers compelling support for a direct association between neuronal ferritinophagy and ferroptosis. Hydroxysafflor Yellow A (HSYA), a traditional Chinese herb, shows promise in reducing brain ischemia/reperfusion injury, but its exact protective mechanism is still unknown. Our study reveals a new way HSYA protects the brain by preventing neuronal ferroptosis after a stroke, a mechanism not previously reported.
Collapse
Affiliation(s)
- Yige Wu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Lijun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Zeqian Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Shuwen Yuan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Dong Ma
- Dept. of Neurosurgery/The Key Laboratory of prevention and treatment of neurological disease of Shanxi Provincial Health Commission, Sinopharm Tongmei General Hospital, Datong 037003, China
| | - Chunli Wen
- Shanxi provincial people's Hospital, Taiyuan 030001, China
| | - Hao Tian
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Baoguo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200040, China.
| | - Cungen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| | - Lijuan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China; Department of Physiology, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
27
|
Ying X, Gao K, Luo Z, Ren Y, Shen C, Zhang H, Zhan C, Xu Z, Wu J, Wu G. Paliperidone Inhibits Ferroptosis Mediated by Autophagy in Renal Tubular Epithelial Cells by Targeting CHAC1. Adv Biol (Weinh) 2025:e2400648. [PMID: 40259582 DOI: 10.1002/adbi.202400648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/18/2025] [Indexed: 04/23/2025]
Abstract
Renal tubular epithelial cell injury is a significant factor in the formation of kidney stones. However, the regulatory mechanisms behind this injury, especially the association with autophagy-mediated ferroptosis, remain unclear. This study first identified the upregulated ferroptosis related gene ChaC Glutathione Specific Gamma-Glutamylcyclotransferase 1 (CHAC1) in kidney stone samples through bioinformatics analysis. Subsequently, a damage model is established by treating renal tubular epithelial cells (HK-2) cells with calcium oxalate (CaOx) and investigated its function by downregulating CHAC1 expression through shRNA transfection. Autophagy status and oxidative stress are evaluated by detecting autophagy (LC3I, LC3II, Beclin 1) and ferroptosis (GPX4) related protein expression using GFP-LC3 adenovirus and Western Blot. In addition, the interaction between small molecule drug Paliperidone (Pali) and CHAC1 is also investigated through molecular docking and cell thermal migration assays to explore therapeutic potential. CHAC1 is upregulated in kidney stones and associated with ferroptosis. Knockdown of CHAC1 weakened CaOx-induced autophagy and ferroptosis. Moreover, Pali can target CHAC1 protein, reduce CHAC1 activity, and inhibit autophagy-mediated ferroptosis during cellular injury. Pali can inhibit autophagy-mediated ferroptosis in renal tubular epithelial cells by targeting CHAC1, offering a new direction for the treatment of kidney stones.
Collapse
Affiliation(s)
- Xiangrong Ying
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Ke Gao
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Zhengang Luo
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Yu Ren
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Chong Shen
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Haojie Zhang
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Chuanchuan Zhan
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Zibin Xu
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Jintao Wu
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| | - Gangfeng Wu
- Department of Urology, Shaoxing People's Hospital, Shaoxing, 312000, China
| |
Collapse
|
28
|
Gupta G, Samuel VP, M RM, Rani B, Sasikumar Y, Nayak PP, Sudan P, Goyal K, Oliver BG, Chakraborty A, Dua K. Caspase-independent cell death in lung cancer: from mechanisms to clinical applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04149-0. [PMID: 40257494 DOI: 10.1007/s00210-025-04149-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/05/2025] [Indexed: 04/22/2025]
Abstract
Caspase-independent cell death (CICD) has recently become a very important mechanism in lung cancer, in particular, to overcome a critical failure in apoptotic cell death that is common to disease progression and treatment failures. The pathways involved in CICD span from necroptosis, ferroptosis, mitochondrial dysfunction, and autophagy-mediated cell death. Its potential therapeutic applications have been recently highlighted. Glutathione peroxidase 4 (GPX4) inhibition-driven ferroptosis has overcome drug resistance in non-small cell lung cancer (NSCLC). In addition, necroptosis involving RIPK1 and RIPK3 causes tumor cell death and modulation of immune responses in the tumor microenvironment (TME). Mitochondrial pathways are critical for CICD through modulation of metabolic and redox homeostasis. Ferroptosis is amplified by mitochondrial reactive oxygen species (ROS) and lipid peroxidation in lung cancer cells, and mitochondrial depolarization induces oxidative stress and leads to cell death. In addition, mitochondria-mediated autophagy, or mitophagy, results in the clearance of damaged organelles under stress conditions, while this function is also linked to CICD when dysregulated. The role of cell death through autophagy regulated by ATG proteins and PI3K/AKT/mTOR pathway is dual: to suppress tumor and to sensitize cells to therapy. A promising approach to enhancing therapeutic outcomes involves targeting mechanisms of CICD, including inducing ferroptosis by SLC7A11 inhibition, modulating mitochondrial ROS generation, or combining inhibition of autophagy with chemotherapy. Here, we review the molecular underpinnings of CICD, particularly on mitochondrial pathways and their potential to transform lung cancer treatment.
Collapse
Affiliation(s)
- Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vijaya Paul Samuel
- Department of Anatomy, RAK College of Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Bindu Rani
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Y Sasikumar
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Priya Priyadarshini Nayak
- Department of Medical Oncology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Puneet Sudan
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Brian G Oliver
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Amlan Chakraborty
- Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, VIC, 3800, Australia
| | - Kamal Dua
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia.
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
29
|
Taghizadieh M, Kalantari M, Bakhshali R, Kobravi S, Khalilollah S, Baghi HB, Bayat M, Nahand JS, Akhavan-Sigari R. To be or not to be: navigating the influence of MicroRNAs on cervical cancer cell death. Cancer Cell Int 2025; 25:153. [PMID: 40251577 PMCID: PMC12008905 DOI: 10.1186/s12935-025-03786-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
With all diagnostic and therapeutic advances, such as surgery, radiation- and chemo-therapy, cervical cancer (CC) is still ranked fourth among the most frequent cancers in women globally. New biomarkers and therapeutic targets are warranted to be discovered for the early detection, treatment, and prognosis of CC. As component of the non-coding RNA's family, microRNAs (miRNAs) participate in several cellular functions such as cell proliferation, gene expression, many signaling cascades, apoptosis, angiogenesis, etc. MiRNAs can suppress or induce programmed cell death (PCD) pathways by altering their regulatory genes. Besides, abnormal expression of miRNAs weakens or promotes various signaling pathways associated with PCD, resulting in the development of human diseases such as CC. For that reason, understanding the effects that miRNAs exert on the various modes of tumor PCD, and evaluating the potential of miRNAs to serve as targets for induction of cell death and reappearance of chemotherapy. The current study aims to define the effect that miRNAs exert on cell apoptosis, autophagy, pyroptosis, ferroptosis, and anoikis in cervical cancer to investigate possible targets for cervical cancer therapy. Manipulating the PCD pathways by miRNAs could be considered a primary therapeutic strategy for cervical cancer.
Collapse
Affiliation(s)
- Mohammad Taghizadieh
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Kalantari
- Department of Biology, Tehran University of health Sciences, Tehran, Iran
| | | | - Sepehr Kobravi
- Department of Oral & Maxillofacial Surgery, Faculty of Dentistry, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Shayan Khalilollah
- Department of Neurosurgery, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
30
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
31
|
Jiang Y, Saeed TN, Alfarttoosi KH, Bishoyi AK, Rekha MM, Kundlas M, Jain B, Rizaev J, Taher WM, Alwan M, Jawad MJ, Ali Al-Nuaimi AM. The intersection of ferroptosis and non-coding RNAs: a novel approach to ovarian cancer. Eur J Med Res 2025; 30:300. [PMID: 40247379 PMCID: PMC12007203 DOI: 10.1186/s40001-025-02559-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
Understanding the core principles of ovarian cancer has been significantly improved through the exploration of Ferroptosis, a type of cell death triggered by iron that leads to an increase in lipid peroxides. Current research has shed light on the critical functions of non-coding RNAs, such as circRNAs, lncRNAs, and miRNAs, in regulating ferroptosis in ovarian cancer. The aim of this paper is to comprehensively analyze how ncRNAs influence the development of ferroptosis in ovarian cancer cells. In-depth exploration is undertaken to understand the intricate ways in which ncRNAs regulate essential elements of ferroptosis, including iron management and lipid peroxidation levels. We also investigate their significant involvement in the progression of this type of cellular demise. It should be emphasized that ncRNAs can impact the synthesis of crucial proteins, such as GPX4, a key contributor to the cellular defense against oxidation, and ACSL4, involved in lipid formation. In addition, we examine the correlation between ncRNAs and well-known pathways associated with oxidative stress and cell death. The consequences of these discoveries are noteworthy, since focusing on particular ncRNAs could potentially render ovarian cancer cells more vulnerable to ferroptosis, effectively combating drug resistance problems. This discussion highlights the growing significance of ncRNAs in governing ferroptosis and their potential as useful biomarkers and treatment targets for ovarian cancer. We intend to promote additional research into the involvement of ncRNAs in controlling ferroptosis, based on current findings, with the ultimate goal of informing targeted therapeutic strategies and improving long-term treatment outcomes for individuals suffering from OC.
Collapse
Affiliation(s)
- Youyi Jiang
- School of Civil Engineering, Chongqing Jiaotong University, Chongqing, China
| | - Tamara Nazar Saeed
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq.
| | | | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Bhavik Jain
- Chitkara Centre for Research and Development, Chitkara University, Baddi, Himachal Pradesh, 174103, India
| | - Jasur Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
32
|
Prata RBDS, Pinheiro RO. Cell Death Mechanisms in Mycobacterium abscessus Infection: A Double-Edged Sword. Pathogens 2025; 14:391. [PMID: 40333197 PMCID: PMC12030298 DOI: 10.3390/pathogens14040391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025] Open
Abstract
Infections caused by non-tuberculous mycobacteria (NTM), such as Mycobacterium abscessus, elicit diverse cell death mechanisms including apoptosis, necrosis, and pyroptosis, which play key roles in immunopathogenesis. NTM can manipulate these cell death pathways to evade host immune responses, ensuring their intracellular survival and persistence. Apoptosis may aid in antigen presentation and immune activation, while necrosis and pyroptosis trigger excessive inflammation, leading to tissue damage. Autophagy, a crucial cellular defense mechanism, is often induced in response to NTM infection; however, M. abscessus has evolved mechanisms to inhibit autophagic processes, enhancing its ability to survive within host cells. This manipulation of cell death pathways, particularly the dysregulation of autophagy and ferroptosis, contributes to chronic infection, immune evasion, and tissue damage, complicating disease management. Understanding these mechanisms offers potential therapeutic targets for improving treatment strategies against M. abscessus infections.
Collapse
Affiliation(s)
| | - Roberta Olmo Pinheiro
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro 21040-360, Brazil;
| |
Collapse
|
33
|
Du Y, Wu H, Zhan S, Zhang R, Zhang G, Bu N. Calcium/Calmodulin-Dependent Protein Kinase II β Regulates Autophagy Dependent Ferroptosis of Neurons after Cerebral Ischemic Injury by Activating the AREG/JUN/ELAVL1 Pathway. Neurochem Res 2025; 50:140. [PMID: 40220216 DOI: 10.1007/s11064-025-04392-0] [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: 09/13/2024] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Ferroptosis is an iron-dependent regulatory cell death characterized by lipid peroxidation. The molecular mechanism of calcium/calmodulin-dependent protein kinase II β (CAMK2B) affecting cerebral ischemic injury through autophagy-dependent ferroptosis is still unclear. Here, we aimed to study the regulatory effect of CAMK2B on autophagy-dependent ferroptosis and its effect on cerebral ischemic injury. We found that CAMK2B was significantly upregulated in oxygen and glucose deprivation/recovery (OGD/R)-induced PC12 cells and primary hippocampal neurons. CAMK2B knockdown inhibited OGD/R-induced autophagy-dependent ferroptosis in PC12 cells and primary hippocampal neurons. In addition, CAMK2B was co-localized with amphiregulin (AREG) in PC12 cells, and overexpression of AREG reversed the effect of CAMK2B knockdown on OGD/R-induced autophagy-dependent ferroptosis in PC12 cells and primary hippocampal neurons. Further molecular mechanism studies showed that AREG enhanced the transcriptional activation of embryonic lethal abnormal vision-like 1 (ELAVL1) through Jun Proto-Oncogene (c-Jun), thereby inducing autophagy-dependent ferroptosis in PC12 cells and primary hippocampal neurons. Moreover, CAMK2B was significantly upregulated in the ipsilateral penumbra neurons of cerebral ischemia-reperfusion (I/R) mice, and the level of autophagy-dependent ferroptosis was increased in the brain tissue of I/R mice. Knockdown of CAMK2B alleviated neuronal damage by inhibiting autophagy-dependent ferroptosis in the brain tissue of model mice. This study suggests that CAMK2B plays a key role in regulating neuronal autophagy-dependent ferroptosis, and CAMK2B may be a potential target for the treatment of cerebral I/R injury.
Collapse
Affiliation(s)
- Yun Du
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xi Wu Road, Xin Cheng District, Xi'an, 710004, Shaanxi Province, China.
| | - Haiqin Wu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xi Wu Road, Xin Cheng District, Xi'an, 710004, Shaanxi Province, China
| | - Shuqin Zhan
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xi Wu Road, Xin Cheng District, Xi'an, 710004, Shaanxi Province, China
| | - Ru Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xi Wu Road, Xin Cheng District, Xi'an, 710004, Shaanxi Province, China
| | - Guilian Zhang
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xi Wu Road, Xin Cheng District, Xi'an, 710004, Shaanxi Province, China
| | - Ning Bu
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, No.157, Xi Wu Road, Xin Cheng District, Xi'an, 710004, Shaanxi Province, China
| |
Collapse
|
34
|
Wang X, Xue Y, Chang L, Zhu X, Liu W, Liang T. The Regulation of Trace Metal Elements in Cancer Ferroptosis. Adv Biol (Weinh) 2025:e2400821. [PMID: 40200790 DOI: 10.1002/adbi.202400821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/11/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, as novel type of regulated cell death that has garnered widespread attention over the past decade, has witnessed the continuous discovery of an increasing number of regulatory mechanisms. Trace metal elements play a multifaceted and crucial role in oncology. Interestingly, it has been increasingly evident that these elements, such as copper, are involved in the regulation of iron accumulation, lipid peroxidation and antiferroptotic systems, suggesting the existence of "nonferrous" mechanisms in ferroptosis. In this review, a comprehensive overview of the composition and mechanism of ferroptosis is provided. The interaction between copper metabolism (including cuproptosis) and ferroptosis in cancer, as well as the roles of other trace metal elements (such as zinc, manganese, cobalt, and molybdenum) in ferroptosis are specifically focused. Furthermore, the applications of nanomaterials based on these metals in cancer therapy are also reviewed and potential strategies for co-targeting ferroptosis and cuproptosis are explored. Nevertheless, in light of the intricate and ambiguous nature of these interactions, ongoing research is essential to further elucidate the "nonferrous" mechanisms of ferroptosis, thereby facilitating the development of novel therapeutic targets and approaches for cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuanyuan Xue
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lei Chang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xuena Zhu
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Wenjun Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
- Zhejiang University Cancer Center, Hangzhou, 310003, China
| |
Collapse
|
35
|
Gong K, Zhou S, Xiao L, Xu M, Zhou Y, Lu K, Yu X, Zhu J, Liu C, Zhu Q. Danggui Shaoyao San ameliorates Alzheimer's disease by regulating lipid metabolism and inhibiting neuronal ferroptosis through the AMPK/Sp1/ACSL4 signaling pathway. Front Pharmacol 2025; 16:1588375. [PMID: 40271063 PMCID: PMC12014676 DOI: 10.3389/fphar.2025.1588375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline; recent studies suggest that neuronal ferroptosis plays a key role in its pathogenesis. Danggui Shaoyao San (DSS), a traditional Chinese medicine formula, has shown demonstrated neuroprotective effects, but its precise mechanisms in AD treatment remain unclear. This study aims to investigate the mechanism of DSS in treating AD by inhibiting neuronal ferroptosis, explore whether DSS alleviates AD by suppressing neuronal ferroptosis via the AMPK/Sp1/ACSL4 pathway. Methods Chemical composition of DSS was identified by LC-MS/MS, followed by network pharmacology to predict targets and pathways. Molecular docking assessed binding affinities between DSS compounds and key proteins (AMPK, Sp1, ACSL4). In vivo experiments on APP/PS1 mice evaluated DSS effects on cognitive function, oxidative stress markers, lipid peroxidation, and ferroptosis-related proteins. Results Network pharmacology analysis suggested that DSS regulates lipid metabolism and inhibits neuronal ferroptosis via the AMPK pathway. Molecular docking revealed strong binding affinities between DSS compounds and AMPK downstream proteins, Sp1 and ACSL4. In vivo experiments showed that DSS improved cognitive function, enhanced antioxidant capacity, reduced lipid peroxide accumulation, and decreased Fe2+ content in brain tissue. Furthermore, DSS increased the expression of FTH, p-AMPK, and GPX4 while decreasing Sp1 and ACSL4 levels, thereby inhibiting ferroptosis. Conclusion DSS alleviates AD symptoms by suppressing neuronal ferroptosis via the AMPK/Sp1/ACSL4 axis, representing a novel lipid metabolism-targeted therapeutic strategy.
Collapse
Affiliation(s)
- Kai Gong
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuang Zhou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Li Xiao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengzhen Xu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuhe Zhou
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kaihui Lu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin Yu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiang Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Jinan, China
| |
Collapse
|
36
|
Wu C, Xu Z, Wang Q, Guo H, He X, Lin Y, Li L, Feng L, Li Q, Tang C. Investigation of Ferroptosis Mechanisms in Ischemic Stroke Treated with Electroacupuncture: Focusing on the NCOA4-FTH1 Signaling Pathway. Neurochem Res 2025; 50:137. [PMID: 40189641 DOI: 10.1007/s11064-025-04390-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025]
Abstract
Ischemic stroke remains a primary cause of mortality and morbidity, with ferroptosis emerging as a critical mechanism underlying neuronal damage post-ischemic injury. This study aims to elucidate the mechanisms of ferroptosis in ischemic stroke and assess the therapeutic potential of electroacupuncture, with emphasis on the NCOA4-FTH1 signaling pathway. After establishing a mouse model of middle cerebral artery occlusion (MCAO), we employed a combination of behavioral assessments and molecular techniques, including transmission electron microscopy, immunofluorescence, and Western blotting, to investigate the impact of electroacupuncture on ferroptosis markers. In addition, we constructed in vivo models of NCOA4 gene silencing and overexpression using adeno-associated virus (AAV) to verify whether electroacupuncture modulates the mechanism of ischemic stroke ferroptosis via the NCOA4-FTH1 signaling pathway. Our findings indicated that electroacupuncture could significantly downregulate NCOA4 expression while upregulating FTH1 and GPX4 levels in affected brain regions of MCAO mice. This resulted in reduced MDA levels, decreased iron ion concentration, a smaller brain infarct area, and improved motor function (p < 0.05). After constructing in vivo models of AAV-mediated NCOA4 gene silencing and overexpression, we demonstrated that electroacupuncture could attenuate iron deposition and inhibit ferroptosis in neurons by suppressing NCOA4 and upregulating FTH1, thereby ameliorating neurological deficits in the ischemic stroke model. These results suggest that electroacupuncture modulates ferroptosis through the NCOA4-FTH1 pathway, offering a novel therapeutic approach for neuroprotection following ischemic stroke.
Collapse
Affiliation(s)
- Chunxiao Wu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518104, P. R. China.
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, Guangdong Province, P. R. China.
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine; Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, P. R. China.
| | - Zhirui Xu
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 510130, Guangdong Province, P. R. China
| | - Qizhang Wang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, 518104, P. R. China
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518104, Guangdong Province, P. R. China
| | - Hongji Guo
- School of Medicine and Health, Shunde Polytechnic, Foshan, 528000, China
| | - Xin He
- Guangzhou Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou, Guangdong Province, P. R. China
| | - Yuexi Lin
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, P. R. China
| | - Luping Li
- Guangzhou City Construction College, Guangzhou, Guangdong Province, P. R. China
| | - Linling Feng
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, P. R. China
| | - Qingyou Li
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, P. R. China
| | - Chunzhi Tang
- Clinical Medical of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, P. R. China
| |
Collapse
|
37
|
Wang T, Liu Q, Wu L, Wang L, Jiang Z, Yue Y, Jiang P, Ji Z, Yin M, Zhang N, Han H. Endoplasmic reticulum stress-autophagy axis is involved in copper-induced ovarian ferroptosis. Free Radic Biol Med 2025; 234:1-18. [PMID: 40194638 DOI: 10.1016/j.freeradbiomed.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/02/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
Copper (Cu) contamination has emerged a global public health problem due to the extensive use of Cu in industrial production and daily life. Reproductive damage resulting from Cu exposure has been particularly evident. Wilson's disease (WD) is a recessive genetic disease characterized by impaired Cu metabolism. Female WD patients have often been associated with reproductive impairment. Ferroptosis, a form of iron-dependent regulated cell death, has been identified as being caused by massive lipid peroxide-mediated membrane damage. However, it remains unclear whether ferroptosis is associated with Cu-induced ovarian damage. In this study, the role of ferroptosis in ovarian damage induced by Cu accumulation and its underlying mechanisms were examined through both in vivo and in vitro experiments. The findings indicated that excessive Cu deposition in the ovaries could lead to follicular atresia and ovulation dysfunction, and trigger ferroptosis in ovarian and granulosa cells (GCs). The mechanism may be related to endoplasmic reticulum (ER) stress mediated by the protein kinase RNA-like ER kinase (PERK) pathway, and hyperactivation of autophagy. In addition, Cu-induced autophagy in GCs was found to increase intracellular iron levels via the ferritinophagy pathway, thereby inducing ferroptosis. We also found that mitochondrial reactive oxygen species (MitoROS) may be an onstream facilitator of Cu-induced ferroptosis via activation of the ER stress-autophagy pathway. Our findings suggested that ferroptosis is associated with Cu-induced ovarian damage and is regulated by the MitoROS-ER stress-autophagy axes. These results might provide insights for developing treatment for WD and other diseases related to Cu exposure.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
| | - Qianzhuo Liu
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
| | - Limin Wu
- Center for Reproduction and Genetics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Luyao Wang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
| | - Zhenzhen Jiang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
| | - Yike Yue
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
| | - Pengyu Jiang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
| | - Zhihui Ji
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
| | - Miaozhu Yin
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
| | - Nian Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China
| | - Hui Han
- Department of Neurology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230031, Anhui Province, China.
| |
Collapse
|
38
|
Yuan D, Guo T, Zhu X, Song W, Nie D, Yu H. Icariside II induces ferroptosis through the down-regulation of SLC7A11 in ovarian cancer. J Ovarian Res 2025; 18:71. [PMID: 40186304 PMCID: PMC11971851 DOI: 10.1186/s13048-025-01650-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/16/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Ovarian cancer (OV) is the leading cause of death among gynecological malignancies. This study aimed to investigate the influence of Icariside II on OV in vitro and in vivo and to elucidate whether Icariside II induces ferroptosis in OV cells by regulating SLC7A11 expression. METHODS SKOV3 cells and OV nude mice were treated with Icariside II, a control-plasmid or an SLC7A11-plasmid. EdU assay, flow cytometry, wound-healing assay, and Transwell assays were used to assess cell proliferation, apoptosis, migration, and invasion respectively. Total iron, Fe2+ levels, and intracellular lipid reactive oxygen species (ROS) stimulation were evaluated in both cells and tissues. Levels of cysteine (Cys), glutathione (GSH), and glutathione peroxidase 4 (GPX4) were also analyzed. Ferroptosis markers, including Ptgs2, Chac1, SLC7A11, and apoptosis-associated genes (Bax and Bcl-2), were detected using qRT-PCR, western blotting, and immunohistochemistry (IHC). SLC7A11 expression in OV was explored using data from The Cancer Genome Atlas (TCGA), and validated with IHC staining. RESULTS Icariside II-induced ferroptosis in OV cells was confirmed by elevated Fe2+ and total iron levels, enhanced lipid ROS levels, higher Ptgs2 and Chac1 mRNA levels, and reduced levels of SLC7A11, Cys, GSH, and GPX4 in both in vitro and in vivo models. These effects were partially reversed by the SLC7A11-plasmid. Moreover, Icariside II suppressed SKOV3 cell proliferation, inhibited cells migration and invasion, and promoted apoptosis by downregulating SLC7A11 expression. Furthermore, we found that SLC7A11 expression was upregulated in OV tissues compared to adjacent non-tumor tissues. CONCLUSION Icariside II induces ferroptosis in OV by downregulating SLC7A11 expression in vitro and in vivo. Our study identified Icariside II as a promising therapeutic agent for the treatment of OV.
Collapse
Affiliation(s)
- Donglan Yuan
- Department of Obstetrics and Gynecology, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China
| | - Ting Guo
- Center for Molecular Medicine, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China
| | - Xiaotong Zhu
- Department of Obstetrics and Gynecology, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China
| | - Weiwei Song
- Department of Traditional Chinese Medicine, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China
| | - Dengyun Nie
- Nanjing University of Traditional Chinese Medicine, Nanjing, 210023, China
| | - Hong Yu
- Department of Pathology, Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, Taizhou, 225300, China.
- Affiliated Taizhou People's Hospital of Nanjing University of Traditional Chinese Medicine, No. 399 Hailing Road, Taizhou, 225300, China.
| |
Collapse
|
39
|
Wei YY, Chen TT, Zhang DW, Zhang Y, Li F, Ding YC, Wang MY, Zhang L, Chen KG, Fei GH. Microplastics exacerbate ferroptosis via mitochondrial reactive oxygen species-mediated autophagy in chronic obstructive pulmonary disease. Autophagy 2025:1-27. [PMID: 40114310 DOI: 10.1080/15548627.2025.2481126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025] Open
Abstract
Microplastics (MPs) induce mitochondrial dysfunction and iron accumulation, contributing to mitochondrial macroautophagy/autophagy and ferroptosis, which has increased susceptibility to the exacerbation of chronic obstructive pulmonary disease (COPD); however, the underlying mechanism remains unclear. We demonstrated that MPs intensified inflammation in COPD by enhancing autophagy-dependent ferroptosis (ADF) in vitro and in vivo. In the lung tissues of patients with COPD, the concentrations of MPs, especially polystyrene microplastics (PS-MPs), were significantly higher than that of the control group, as detected by pyrolysis gas chromatography mass spectrometry (Py-GCMS), with increased iron accumulation. The exposure to PS-MPs, 2 μm in size, resulted in their being deposited in the lungs of COPD model mice detected by optical in vivo imaging, and observed in bronchial epithelial cells traced by GFP-labeled PS-MPs. There were mitochondrial impairments accompanied by mitochondrial reactive oxygen species (mito-ROS) overproduction and significantly increased levels of lysosome biogenesis and acidification in pDHBE cells with PS-MP stimulation, triggering occurrence of ferritinophagy and enhancing ADF in COPD, which triggered acute exacerbation of COPD (AECOPD). Reestablishing autophagy-dependent ferroptosis via mitochondria-specific ROS scavenging or ferroptosis inhibition alleviated excessive inflammation and ameliorated AECOPD induced by PS-MPs. Collectively, our data initially revealed that MPs exacerbate ferroptosis via mito-ROS-mediated autophagy in COPD, which sheds light on further hazard assessments of MPs on human respiratory health and potential therapeutic agents for patients with COPD.Abbreviations: ADF: autophagy-dependent ferroptosis; AECOPD: acute exacerbation of chronic obstructive pulmonary disease; Cchord: static compliance; COPD: chronic obstructive pulmonary disease; CQ: chloroquine; CS: cigarette smoke; DEGs: differentially expressed genes; Fer-1: ferrostatin-1; FEV 0.1: forced expiratory volume in first 100 ms; FVC: forced vital capacity; GSH: glutathione; HE: hematoxylin and eosin; IL1B/IL-1β: interleukin 1 beta; IL6: interleukin 6; MDA: malondialdehyde; Mito-ROS: mitochondrial reactive oxygen species; MMA: methyl methacrylate; MMF: maximal mid-expiratory flow curve; MMP: mitochondrial membrane potential; MOI: multiplicity of infection; MPs: microplastics; MV: minute volume; PA: polyamide; PBS: phosphate-buffered saline; PC: polycarbonate; pDHBE: primary human bronchial epithelial cell from COPD patients; PET: polyethylene terephthalate; PIF: peak inspiratory flow; PLA: polylactic acid; pNHBE: primary normal human bronchial epithelial cell; PS-MPs: polystyrene microplastics; PVA: polyvinyl acetate; PVC: polyvinyl chloride; Py-GCMS: pyrolysis gas chromatography mass spectrometry; SEM: scanning electron microscopy; Te: expiratory times; Ti: inspiratory times; TNF/TNF-α: tumor necrosis factor.
Collapse
Affiliation(s)
- Yuan Yuan Wei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Ting Ting Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Da Wei Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Ying Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Fang Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Yi Chuan Ding
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Ming Yu Wang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| | - Ling Zhang
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Ke Gong Chen
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
- Department of Thoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Guang He Fei
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Respiratory Diseases Research and Medical Transformation of Anhui Province, Hefei, Anhui, P.R. China
| |
Collapse
|
40
|
Wang L, Tang C, Zhang Q, Pan Q. Ferroptosis as a molecular target of epigallocatechin gallate in diseases. Arch Physiol Biochem 2025; 131:156-168. [PMID: 39264116 DOI: 10.1080/13813455.2024.2401892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/24/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
CONTEXT Ferroptosis is a novel form of cell death characterised by iron overload and lipid peroxidation. It is closely associated with many diseases, including cardiovascular diseases, tumours, and neurological diseases. The use of natural chemicals to modulate ferroptosis is of great concern because of the critical role ferroptosis plays in disease. The main active ingredient in green tea is epigallocatechin gallate (EGCG), which is the most abundant catechin in green tea. EGCG shows a wide range of biological and therapeutic effects in various diseases, including anti-inflammatory, antioxidant, anticancer, and cardioprotective. OBJECTIVE The purpose of this article is to summarise the existing information on the relationship between EGCG and ferroptosis. METHODS Articles related to EGCG and ferroptosis were searched in PubMed and Web of Science databases, and the literature was analysed. RESULTS AND CONCLUSION EGCG could improve ferroptosis-related diseases and affect the development of ferroptosis by regulating the nuclear factor erythroid 2-related factor 2, autophagy, microRNA, signal transducer and activator of transcription 1, and protein kinase D1 signalling pathways.
Collapse
Affiliation(s)
- Lili Wang
- Wuhan Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Chunlian Tang
- Wuhan Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Qizhi Zhang
- Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Qun Pan
- Wuhan Wuchang Hospital, Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Zeng Y, Duan T, Huang J, Wang X. Astragaloside IV inhibits nasopharyngeal carcinoma progression by suppressing the SATB2/Wnt signaling axis. Toxicol Res (Camb) 2025; 14:tfaf047. [PMID: 40177383 PMCID: PMC11964083 DOI: 10.1093/toxres/tfaf047] [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: 10/11/2024] [Revised: 02/10/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Astragaloside IV (AS-IV), a major bioactive component of Astragalus membranaceus, exhibits anti-cancer and anti-inflammatory properties. However, its precise role in nasopharyngeal carcinoma (NPC) remains unclear. This study investigated the effects of AS-IV on NPC progression and its relationship with Special AT-rich binding protein-2 (SATB2), a diagnostic marker for NPC. AS-IV treatment reduced NPC cell viability in a dose-dependent manner, as assessed by CCK-8 assays. Functional experiments, including transwell, immunofluorescence, and flow cytometry assays, demonstrated that AS-IV inhibited cell migration, invasion, and autophagy while promoting apoptosis. Western blot analysis showed that SATB2 expression was significantly elevated in NPC cells, particularly in C666-1 and HK-1 cells. Overexpression of SATB2 partially reversed AS-IV's inhibitory effects on NPC progression. Further analysis revealed that AS-IV suppressed the Wnt signaling pathway by downregulating SATB2 expression, while SATB2 overexpression restored Wnt pathway activation. This effect was reversed upon treatment with the Wnt pathway inhibitor DKK-1. In vivo, AS-IV administration inhibited tumor growth in a nude mouse subcutaneous xenograft model, reduced Ki-67 positivity, and lowered LC3B expression, indicating decreased proliferation and autophagy. However, these effects were diminished upon SATB2 overexpression. These findings suggest that AS-IV exerts anti-tumor effects in NPC by downregulating SATB2 and suppressing Wnt pathway activation, highlighting its potential as a therapeutic agent for NPC. Highlights Astragaloside IV (AS-IV) reduces nasopharyngeal carcinoma (NPC) cell vitality, suppresses cell migration, invasion and autophagy, and fosters apoptosis.SATB2 exhibits notably high levels in NPC cells.Overexpression of SATB2 counteracts the inhibition of NPC malignant progression by AS-IV.AS-IV impedes NPC progression by decreasing SATB2 and thereby hindering the Wnt pathway.AS-IV deters NPC tumor growth in nude mice.
Collapse
Affiliation(s)
- Yinping Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Tingting Duan
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Jiajun Huang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| | - Xiaofeng Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Longhua District, Haikou 570102, Hainan Province, China
| |
Collapse
|
42
|
Han S, Zhao X, Yu C, Cui C, Zhang Y, Zhu Q, Qiu M, Yang C, Yin H. Nestin Regulates Autophagy-Dependent Ferroptosis Mediated Skeletal Muscle Atrophy by Ubiquitinating MAP 1LC3B. J Cachexia Sarcopenia Muscle 2025; 16:e13779. [PMID: 40183241 PMCID: PMC11969254 DOI: 10.1002/jcsm.13779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Programmed cell death plays a critical role in skeletal muscle atrophy. Ferroptosis, an iron-dependent form of programmed cell death driven by lipid peroxidation, has been implicated in various diseases, but its role in skeletal muscle atrophy remains unclear. METHODS Ferroptosis in skeletal muscle atrophy was investigated using two models: dexamethasone (Dex)-induced atrophy (n = 6 independent cell cultures per group) and simulated microgravity (n = 6 mice per group). Conditional Nestin knockout (KO) mice were generated using CRISPR/Cas9 (n = 6-8 mice per group), with wild-type (WT) controls (n = 6-8). Phenotypic analyses included histopathology (HE staining), functional assessments (muscle strength, weight analysis, treadmill), and dystrophy evaluation (dystrophin staining). Molecular analyses involved flow cytometry, ELISA, transmission electron microscopy, PI staining, and IP/MS to delineate Nestin-regulated ferroptosis pathways in skeletal muscle atrophy. RESULTS Ferroptosis was significantly activated in both atrophy models, with a 2.5-fold increase in lipid peroxidation (p < 0.01), a 2-fold accumulation of Fe2+ (p < 0.01) and a 50% reduction in Nestin expression (p < 0.001). Nestin KO mice exhibited exacerbated muscle atrophy, showing a 40% decrease in muscle weight (p < 0.01) and a 30% reduction in muscle strength (p < 0.05) compared to WT mice. Nestin overexpression mitigated Dex-induced ferroptosis, reducing lipid peroxidation by 40%, decreasing Fe2+ accumulation by 50% (p < 0.01), and improving muscle function by 30% (p < 0.05). Mechanistically, Nestin interacted with MAP 1LC3B (LC3B) to catalyse LC3B polyubiquitination at lysine-51, reducing LC3B availability for autophagy and inhibiting autophagy flux by 60% (p < 0.01), leading to a 50% reduction in ferroptosis (p < 0.001). CONCLUSIONS Our study identifies Nestin as a critical regulator of ferroptosis-autophagy crosstalk in skeletal muscle atrophy. Targeting Nestin-LC3B ubiquitination may offer novel therapeutic strategies for preventing muscle wasting in diseases such as cachexia and sarcopenia.
Collapse
Affiliation(s)
- Shunshun Han
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Xiyu Zhao
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Chunlin Yu
- Animal Breeding and Genetics Key Laboratory of Sichuan ProvinceSichuan Animal Science AcademyChengduChina
| | - Can Cui
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Yao Zhang
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Qing Zhu
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| | - Mohan Qiu
- Animal Breeding and Genetics Key Laboratory of Sichuan ProvinceSichuan Animal Science AcademyChengduChina
| | - Chaowu Yang
- Animal Breeding and Genetics Key Laboratory of Sichuan ProvinceSichuan Animal Science AcademyChengduChina
| | - Huadong Yin
- College of Animal Science and TechnologyKey Laboratory of Livestock and Poultry Multi‐Omics, Ministry of Agriculture and Rural AffairsSichuan Agricultural UniversityChengduSichuanChina
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan ProvinceSichuan Agricultural UniversityChengduSichuanChina
| |
Collapse
|
43
|
Yuan F, Han S, Li Y, Li S, Li D, Tian Q, Feng R, Shao Y, Liang X, Wang J, Lei H, Li X, Duan Y. miR-214-3p attenuates ferroptosis-induced cellular damage in a mouse model of diabetic retinopathy through the p53/SLC7A11/GPX4 axis. Exp Eye Res 2025; 253:110299. [PMID: 39978746 DOI: 10.1016/j.exer.2025.110299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Ferroptosis has been implicated in the development of diabetic retinopathy (DR). This study aimed to identify novel ferroptosis-related regulators involved in the pathophysiology of DR using an in vivo streptozotocin (STZ)-induced diabetic model in C57BL/6J mice and cultured primary human retinal vascular endothelial cells (HRECs). Transmission electron microscopy revealed mitochondrial morphological changes consistent with ferroptosis in vascular endothelial cells from STZ-treated mice. Western blot analysis showed increased levels of ferroptosis markers (4-HNE, p53, phosphorylated p53) along with decreased levels of glutathione (GSH), SLC7A11, and GPX4 in diabetic mice. In vitro experiments demonstrated that ferroptosis inhibitors, including pifithrin-α (a p53 inhibitor) and ferrostatin-1 (Fer-1), mitigated cellular damage and Fe2+ accumulation in high-glucose-treated HRECs. These inhibitors also improved mitochondrial membrane potential and restored GSH levels. Bioinformatics analysis and dual-luciferase assays identified a p53 binding site within the miR-214-3p sequence. Overexpression of miR-214-3p in high-glucose-treated HRECs resulted in downregulation of p53 and upregulation of SLC7A11 and GPX4, thereby alleviating ferroptosis-induced injury. This study demonstrates that ferroptosis contributes to retinal damage at tissue, cellular, and molecular levels in DR. Specifically, p53, regulated by miR-214-3p, promotes ferroptosis through the SLC7A11/GPX4 pathway under high-glucose conditions. These findings suggest that the miR-214-3p/p53/SLC7A11/GPX4 axis could serve as a potential therapeutic target for managing ferroptosis and retinal damage in diabetic retinopathy.
Collapse
Affiliation(s)
- Fang Yuan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China; Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Songyu Han
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Yahong Li
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sanming Li
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Dian Li
- Washington University in St. Louis, MO, USA
| | - Qingjun Tian
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Ronghua Feng
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ying Shao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xing Liang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingbo Wang
- Shanxi Eye Hospital Affiliated with Shanxi Medical University, Taiyuan, 030072, China
| | - Hetian Lei
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.
| | - Yajian Duan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China; Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
44
|
Wang G, Li Z, Han W, Tian Q, Liu C, Jiang S, Xiang X, Zhao X, Wang L, Liao J, Li M. Itaconate promotes mitophagy to inhibit neuronal ferroptosis after subarachnoid hemorrhage. Apoptosis 2025; 30:991-1004. [PMID: 39924585 DOI: 10.1007/s10495-025-02077-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2025] [Indexed: 02/11/2025]
Abstract
Subarachnoid hemorrhage (SAH), representing 5-10% of all stroke cases, is a cerebrovascular event associated with a high mortality rate and a challenging prognosis. The role of IRG1-regulated itaconate in bridging metabolism, inflammation, oxidative stress, and immune response is pivotal; however, its implications in the early brain injury following SAH remain elusive. The SAH nerve inflammation model was constructed by Hemin solution and BV2 cells. In vitro and in vivo SAH models were established by intravascular puncture and Hemin solution treatment of HT22 cells. To explore the relationship between IRG1 and neuroinflammation by interfering the expression of Irg1 in BV2 cells. By adding itaconate and its derivatives to explore the relationship between mitophagy and ferroptosis. IRG1 knockdown increased the expression of inflammatory factors and induced the transformation of microglia to pro-inflammatory phenotype after SAH; Itaconate and itaconate derivative 4-OI can reduce oxidative stress and lipid peroxidation level in neuron after SAH, and reduce EBI after SAH; IRG1/ itaconate promotes mitophagy through PINK1/Parkin signaling pathway to inhibit neuronal ferroptosis. IRG1 can improve nerve inflammation after SAH, M2 of microglia induced polarization. IRG1/ Itaconate participates in mitophagy through PINK1/Parkin to alleviate neuronal ferroptosis after SAH and play a neuroprotective role.
Collapse
Affiliation(s)
- Guijun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Zhijie Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Wenrui Han
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Chengli Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Shengming Jiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Xi Xiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Xincan Zhao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Lei Wang
- The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China.
- Department of Neurosurgery, Yichang Central People's Hospital, Yichang, Hubei, China.
| | - Jianming Liao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China.
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China.
- 99 Ziyang Road, Wuhan, Hubei Province, 430060, China.
| |
Collapse
|
45
|
Hong X, Liu H, Sun H, Zhuang Y, Xiao M, Li S, Li Y, Jing M. Yunnan medicine Jiangzhi ointment alleviates hyperlipid-induced hepatocyte ferroptosis by activating AMPK and promoting autophagy. Cytotechnology 2025; 77:73. [PMID: 40062225 PMCID: PMC11883071 DOI: 10.1007/s10616-025-00737-3] [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: 12/04/2024] [Accepted: 02/27/2025] [Indexed: 03/21/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a serious public health problem worldwide. The purpose of this study was to investigate whether Yunnan medicine Jiangzhi ointment (YMJO) can relieve the progression of NAFLD and to elucidate the specific mechanism involved. A NAFLD model was established in high-fat diet (HFD)-induced SD rats and free fatty acid (FFA)-induced BRL 3A cells. The expression of autophagy-related proteins and ferroptosis-related proteins was detected using Western blotting. The histopathological features of the livers of NAFLD rats were evaluated using hematoxylin and eosin (HE) and Oil Red O staining. The results revealed that in a successfully established HFD-induced NAFLD rat model, YMJO alleviated the progression of NAFLD, promoted autophagy, and inhibited ferroptosis. This regulatory mechanism is related to the activation of the AMPK pathway. Further study of the molecular mechanism via cell experiments revealed that YMJO activated FFA-induced liver cell autophagy through the AMPK signaling pathway and inhibited ferroptosis, thus alleviating the development of NAFLD. This study revealed that YMJO promotes phosphorylation by activating the AMPK pathway, enhances autophagy, ameliorates ferroptosis induced by high fat, and alleviates the occurrence and development of NAFLD.
Collapse
Affiliation(s)
- Xin Hong
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Haijing Liu
- Department of Acupuncture and Massage, Yunnan University of Chinese Medicine, Kunming, 650500 China
| | - Hongli Sun
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Yan Zhuang
- Department of Pharmaceutical Preparation, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Meizhen Xiao
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Shaoping Li
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Yandong Li
- Department of Preventive Medicine, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| | - Ming Jing
- Department of Acupuncture and Massage, Yuxi Municipal Hospital of TCM, Yuxi, 653100 China
| |
Collapse
|
46
|
Huang S, Ji P, Xu P, Liu K, Ge H, Yan Z, Cheng Q, Lv J, Zhang D. PLAGL2-STAU1-NCOA4 axis enhances gastric cancer peritoneal metastasis by resisting ferroptosis via ferritinophagy. Apoptosis 2025; 30:1058-1075. [PMID: 39987411 DOI: 10.1007/s10495-025-02083-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/24/2025]
Abstract
Peritoneal metastasis (PM) is the primary site of distant metastasis in gastric cancer (GC) and is associated with an advanced disease stage and poor prognosis. Due to its high resistance to chemotherapy, disseminated peritoneal lesions are often untreatable. A primary reason for therapy resistance in cancer cells is often their defective cell death execution mechanisms. Ferroptosis, a newly identified type of regulated cell death, is strongly linked to the emergence and formation of tumors. Earlier studies have demonstrated the significant role of RNA-binding proteins in ferroptosis. Nevertheless, the fundamental process linking Staufen Double-Stranded RNA Binding Protein 1 (STAU1) to ferroptosis in the peritoneal metastasis of gastric cancer is yet to be clarified. This study shows that the RNA-binding protein STAU1 is crucial for regulating ferroptosis in gastric cancer cells. Elevated levels of STAU1 are linked to unfavorable outcomes in individuals diagnosed with gastric cancer. STAU1 was up-regulated by PLAGL2 and decreased the stability of NCOA4 mRNA by binding to the 3'-untranslated region. Decreased NCOA4 expression inhibits the accumulation of reactive iron, the occurrence of the Fenton reaction, and cellular ROS generation in the GC cells. Additionally, we showed that NCOA4 is crucial in the process of ferritinophagy triggered by the reduction of STAU1 in gastric cancer cells. Ultimately, the process safeguards GC cells from ferroptosis. These findings elucidate the function of PLAGL2/STAU1/NCOA4 in the ferroptosis of gastric cancer cells and provide theoretical backing for possible diagnostic markers and treatment targets for peritoneal metastasis in gastric cancer.
Collapse
Affiliation(s)
- Shansong Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Peicheng Ji
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Peng Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kanghui Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Han Ge
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Zhengyuan Yan
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of Surgery, Nanjing Lishui People's Hospital, Nanjing, 211200, China
| | - Quan Cheng
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jialun Lv
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Department of General Surgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, Jiangsu Province, China.
| |
Collapse
|
47
|
Min L, Huo F, Zhu Z, Din L, Zhang L, Xu Y, Xing X, Zhang P, Wang Q. Mechanistic study of METTL3 inducing ferroptosis to promote cervical cancer progression through mediating m6A modification of COTE-1. Cell Signal 2025; 128:111649. [PMID: 39923928 DOI: 10.1016/j.cellsig.2025.111649] [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: 01/02/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Cervical Cancer (CC) is one of the leading causes of tumor-related deaths among women worldwide, and the mechanisms underlying the anti-ferroptosis of CC cells are still unclear. Methyltransferase like 3 (METTL3) is widely expressed various types of tissues and plays a crucial role in tumorigenesis in part by mediating cell death. However, its regulatory function in CC progression and especially the underlying mechanisms have not been fully elucidated. This study aims to explore the role of METTL3 in the ferroptosis of CC cells. Mechanistically, by MeRIP-seq, we identified COTE-1 as a target of METTL3 mediated m6A modification, and revealed that METTL3-mediated COTE-1 expression was dependent on the m6A reader-dependent manner. Functionally, in vitro and in vivo experiments that METTL3 promotes proliferation and metastasis of CC cells by regulating COTE-1 expression. In addition, the study verified the effect of the METTL3/COTE-1 axis on autophagy-dependent ferroptosis. In summary, METTL3 influences CC progression by mediating COTE-1 to influence autophagy-dependent ferroptosis, representing a potential therapeutic approach for treating CC.
Collapse
Affiliation(s)
- Luyao Min
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Fuchun Huo
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Zhiman Zhu
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lina Din
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lin Zhang
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Yuting Xu
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Xuewei Xing
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Peng Zhang
- Jiangsu Province Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Qingling Wang
- Department of Pathology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
48
|
Yang X, Wu H, Liu D, Zhou G, Zhang D, Yang Q, Liu Y, Li Y. The link between ferroptosis and autophagy in myocardial ischemia/reperfusion injury: new directions for therapy. J Cardiovasc Transl Res 2025; 18:408-423. [PMID: 39885084 DOI: 10.1007/s12265-025-10590-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/10/2025] [Indexed: 02/01/2025]
Abstract
Myocardial ischemia/reperfusion (I/R)-induced cell death, such as autophagy and ferroptosis, is a major contributor to cardiac injury. Regulating cell death may be key to mitigating myocardial ischemia/reperfusion injury (MI/RI). Autophagy is a crucial physiological process involving cellular self-digestion and compensation, responsible for degrading excess or malfunctioning long-lived proteins and organelles. During MI/RI, autophagy plays both "survival" and "death" roles. A growing body of research indicates that ferroptosis is a type of autophagy-dependent cell death. This article provides a comprehensive review of the functions of autophagy and ferroptosis in MI/RI, as well as the molecules mediating their interaction. Understanding the link between autophagy and ferroptosis may offer new therapeutic directions for MI/RI, bearing significant clinical implications.
Collapse
Affiliation(s)
- Xiaoting Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| | - Hui Wu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China.
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China.
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China.
| | - Di Liu
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huang Shi, HuBei Province, China
| | - Gang Zhou
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Dong Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Qingzhuo Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yanfang Liu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| | - Yi Li
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
- Central Laboratory, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, HuBei Province, China
| |
Collapse
|
49
|
Zhuo B, Qin C, Deng S, Jiang H, Si S, Tao F, Cai F, Meng Z. The role of ACSL4 in stroke: mechanisms and potential therapeutic target. Mol Cell Biochem 2025; 480:2223-2246. [PMID: 39496916 PMCID: PMC11961533 DOI: 10.1007/s11010-024-05150-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/24/2024] [Indexed: 11/06/2024]
Abstract
Stroke, as a neurological disorder with a poor overall prognosis, has long plagued the patients. Current stroke therapy lacks effective treatments. Ferroptosis has emerged as a prominent subject of discourse across various maladies in recent years. As an emerging therapeutic target, notwithstanding its initial identification in tumor cells associated with brain diseases, it has lately been recognized as a pivotal factor in the pathological progression of stroke. Acyl-CoA synthetase long-chain family member 4 (ACSL4) is a potential target and biomarker of catalytic unsaturated fatty acids mediating ferroptosis in stroke. Specifically, the upregulation of ACSL4 leads to heightened accumulation of lipid peroxidation products and reactive oxygen species (ROS), thereby exacerbating the progression of ferroptosis in neuronal cells. ACSL4 is present in various tissues and involved in multiple pathways of ferroptosis. At present, the pharmacological mechanisms of targeting ACSL4 to inhibit ferroptosis have been found in many drugs, but the molecular mechanisms of targeting ACSL4 are still in the exploratory stage. This paper introduces the physiopathological mechanism of ACSL4 and the current status of the research involved in ferroptosis crosstalk and epigenetics, and summarizes the application status of ACSL4 in modern pharmacology research, and discusses the potential application value of ACSL4 in the field of stroke.
Collapse
Affiliation(s)
- Bifang Zhuo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chenyang Qin
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shizhe Deng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Hailun Jiang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shangkun Si
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng Tao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Fei Cai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Zhihong Meng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
50
|
Peng X, Yang R, Wang C, Peng W, Zhao Z, Shi S, Cai Q, He B, Wang L, Yu F, Wang X, Tao Y. The YTHDF3-DT/miR-301a-3p /INHBA axis attenuates autophagy-dependent ferroptosis in lung adenocarcinoma. Cancer Lett 2025; 613:217503. [PMID: 39892700 DOI: 10.1016/j.canlet.2025.217503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
YTHDF3-DT, a long non-coding RNA (lncRNA) significantly upregulated in lung adenocarcinoma (LUAD), is associated with poor patient prognosis and plays critical roles in LUAD progression. Clinical data and in vitro analyses revealed that YTHDF3-DT expression correlates with worse overall survival and increased lymph node metastasis in LUAD patients. Functional studies demonstrated that YTHDF3-DT activates the TGF-β and PI3K/Akt/mTOR signaling pathways via INHBA, a key target influenced by YTHDF3-DT. Mechanistically, YTHDF3-DT stabilizes INHBA mRNA by acting as a competing endogenous RNA (ceRNA) for miR-301a-3p, forming a YTHDF3-DT/miR-301a-3p/INHBA axis. This axis regulates ferroptosis in an autophagy-dependent manner in LUAD cells, with YTHDF3-DT promoting cell survival by altering autophagic activity and mitigating ferroptosis-induced cell death. In vivo experiments further validated the role of YTHDF3-DT in tumor growth and ferroptosis regulation, highlighting its potential as a therapeutic target in LUAD. Our data contribute toward a significant mechanistic understanding of the molecules involved in the crosstalk between ferroptosis and autophagy, providing potential therapeutic targets to complement the existing therapies for overcoming the developed resistance in patients with LUAD.
Collapse
Affiliation(s)
- Xiong Peng
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Rui Yang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Department of Pathology, School of Basic Medicine and Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
| | - Christopher Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Weilin Peng
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Zhenyu Zhao
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Shuai Shi
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Qidong Cai
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Boxue He
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Li Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Fenglei Yu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Xiang Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China.
| | - Yongguang Tao
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, 410011, Changsha, Hunan, China; Department of Pathology, Xiangya Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Central South University, 410008, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078, Changsha, Hunan, China.
| |
Collapse
|