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
|
Liu C, Tian Q, Li Z, Wang G, Han W, Jiang S, Sun Z, Xu Q, Wang L, Liao J, Li M. FOXO3a-BAP1 axis regulates neuronal ferroptosis in early brain injury after subarachnoid hemorrhage. Redox Biol 2025; 82:103550. [PMID: 40080966 PMCID: PMC11946873 DOI: 10.1016/j.redox.2025.103550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/15/2025] Open
Abstract
Subarachnoid hemorrhage (SAH) is a serious and common disease and accounts for about 10 % of acute stroke cases. BRCA-associated protein 1 (BAP1) belongs to the ubiquitin C-terminal hydrolases (UCHs) family, which plays an important role in cell metabolism and cell death, but its role in early brain injury (EBI) after SAH requires further study. Forkhead box protein O3a (FOXO3a) is a transcription factor involved in the regulation of cellular function and survival in the nervous system, including the oxidative stress response and neuronal death. This study aimed to explore the effect of FOXO3a and BAP1 on neuronal ferroptosis in the pathogenesis of EBI after SAH. In this study, the overexpression of BAP1 significantly inhibited GPX4 expression and exacerbated the degree of lipid peroxidation and ferroptosis in neurons after SAH. BAP1 regulated the transcription level of the SLC7A11 promoter by H2Aub. FOXO3a could transcriptionally regulate BAP1 to influence the levels of SLC7A11 and GPX4, and mediate lipid peroxidation and neuronal ferroptosis after SAH. Silencing FOXO3 and BAP1 significantly improved neurological deficit and cerebral edema, and reduced oxidative stress damage in SAH mice. After SAH, BAP1 could directly bind to the FKH-DBD + NLS domain located in FOXO3a protein through the UCH domain, and mediates deubiquitination of FOXO3a protein by the K48 site to maintain the stability of FOXO3a. Our findings elucidate the impact of FOXO3a and BAP1 on SLC7A11-related ferroptosis following SAH both in vivo and in vitro, and the inhibition of the FOXO3a-BAP1 axis can significantly attenuate neuronal damage and ferroptosis in EBI after SAH.
Collapse
Affiliation(s)
- Chengli Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Zhijie Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Guijun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Wenrui Han
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Shengming Jiang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Zhou Sun
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Qingqing Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Long Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Jianming Liao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China.
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China.
| |
Collapse
|
3
|
Song Y, Luo X, Yao L, Chen Y, Mao X. Exploring the Role of Ferroptosis-Related Circular RNAs in Subarachnoid Hemorrhage. Mol Biotechnol 2025; 67:1310-1320. [PMID: 38619799 DOI: 10.1007/s12033-024-01140-7] [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: 01/07/2024] [Accepted: 03/06/2024] [Indexed: 04/16/2024]
Abstract
Subarachnoid hemorrhage (SAH) is a devastating cerebrovascular event associated with high mortality and significant morbidity. Recent studies have highlighted the emerging role of ferroptosis, a novel form of regulated cell death, in the pathogenesis of SAH. Circular RNAs (circRNAs), have been found to play essential roles in various cellular processes, including gene regulation and disease pathogenesis. The expression profile of circRNAs in neural tissues, particularly in the brain, suggests their critical role in synaptic function and neurogenesis. Moreover, the interplay between circRNAs and ferroptosis-related pathways, such as iron metabolism and lipid peroxidation, is explored in the context of SAH. Understanding the functional roles of specific circRNAs in the context of SAH may provide potential therapeutic targets to attenuate ferroptosis-associated brain injury. Furthermore, the potential of circRNAs as diagnostic biomarkers for SAH severity, prognosis, and treatment response is discussed. Overall, this review highlights the significance of studying the intricate interplay between circRNAs and ferroptosis in the context of SAH. Unraveling the mechanisms by which circRNAs modulate ferroptotic cell death may pave the way for the development of novel therapeutic strategies and diagnostic approaches for SAH management, ultimately improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Yanju Song
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China
| | - Xin Luo
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China
| | - Liping Yao
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China
| | - Yinchao Chen
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China
| | - Xinfa Mao
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410015, China.
| |
Collapse
|
4
|
Ma J, Zhong X, Li Z, Jiang Y, Jiang Y, Liu X, Hu Y, Yang Z, Zhai G. Di-Dang-Tang suppresses ferroptosis in the hippocampal CA1 region by targeting PGK1/NRF2/GPX4 signaling pathway to exert neuroprotection in vascular dementia. Int Immunopharmacol 2025; 150:114233. [PMID: 39946772 DOI: 10.1016/j.intimp.2025.114233] [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: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 03/03/2025]
Abstract
Increasing evidence has emphasized the crucial role of ferroptosis in the pathogenesis of Vascular dementia (VaD). Di-Dang-Tang (DDT) has the effects of removing blood stasis according to the theory of Traditional Chinese medicine (TCM), while its effects on ferroptosis and mechanisms remain unclear. To elucidate whether the neuroprotective effect of DDT treatment is associated with ferroptosis mediated by the Phosphoglycerate kinase 1 (PGK1)/ Nuclear Factor Erythroid 2-related factor (NRF2)/ Glutathione Peroxidase 4 (GPX4) signaling pathway in the hippocampal CA1 region of rats with the 2-vessel occlusion (2VO) model, we conducted a series of experiments. Nissl staining, HE staining and FJB staining were used to assess the effects of DDT on the degeneration and apoptosis of neurons in the CA1 region of the hippocampus. DDT's suppression on ferroptosis and its protective effects were also evaluated by ELISA and DHE fluorescence. Immunofluorescence assay, immunohistochemistry examination, and western blot analysis further validated DDT's regulatory effects on ferroptosis via PGK1/NRF2/GPX4 pathway. Additionally, we explored the key mediating role of PGK1 in the DDT treatment of VaD by overexpressing PGK1 using AAV-OE-PGK1 plasmid injection. DDT significantly attenuated neuronal apoptosis and degeneration in CA1 region and ameliorated cognitive dysfunctions in VaD rats. DDT inhibited ferroptosis in this brain region, as evidenced by an up-regulation of GPX4 and SLC7A11, and a decline in ferroptosis-related indices. Further, DDT activated protein expression of the PGK1/NRF2/GPX4 pathway, alleviating the lipid peroxidation. Notably, the inhibition of ferroptosis by DDT was achieved by suppression of the PGK1 axis signaling pathway.
Collapse
MESH Headings
- Animals
- Ferroptosis/drug effects
- NF-E2-Related Factor 2/metabolism
- Dementia, Vascular/drug therapy
- Dementia, Vascular/metabolism
- Dementia, Vascular/pathology
- Signal Transduction/drug effects
- Male
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Phosphoglycerate Kinase/metabolism
- Phosphoglycerate Kinase/genetics
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Rats
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Rats, Sprague-Dawley
- Disease Models, Animal
Collapse
Affiliation(s)
- Junjie Ma
- School of Intergrative Medicine, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Qixia District, Nanjing City, Jiangsu 210000, China
| | - Xinxin Zhong
- School of Intergrative Medicine, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Qixia District, Nanjing City, Jiangsu 210000, China
| | - Zhiyuan Li
- School of Intergrative Medicine, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Qixia District, Nanjing City, Jiangsu 210000, China
| | - Yongxia Jiang
- Lianyungang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang City, Jiangsu 222004, China
| | - Yongqu Jiang
- Lianyungang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang City, Jiangsu 222004, China
| | - Xiaoli Liu
- Lianyungang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang City, Jiangsu 222004, China
| | - Yue Hu
- School of Intergrative Medicine, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Qixia District, Nanjing City, Jiangsu 210000, China
| | - Zhou Yang
- Lianyungang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang City, Jiangsu 222004, China
| | - Guojie Zhai
- Department of Neurology, Suzhou Ninth People's Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, No. 2666, Ludang Road, Wujiang District, Suzhou City, Jiangsu 215200, China.
| |
Collapse
|
5
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Wu JW, Wang BX, Shen LP, Chen YL, Du ZY, Du SQ, Lu XJ, Zhao XD. Investigating the Potential Therapeutic Targeting of the JAK-STAT Pathway in Cerebrovascular Diseases: Opportunities and Challenges. Mol Neurobiol 2025:10.1007/s12035-025-04834-4. [PMID: 40102347 DOI: 10.1007/s12035-025-04834-4] [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: 08/02/2024] [Accepted: 03/09/2025] [Indexed: 03/20/2025]
Abstract
Cerebrovascular disease (CVD) is a significant neurological condition resulting from pathological changes in the brain's blood supply and is currently the leading cause of death and disability worldwide. The progression of CVD is closely associated with endothelial damage, plaque formation, and thrombosis, driven by long-term alterations in vascular endothelial cells, smooth muscle cells, microglia, and other immune-inflammatory cells. Among the key molecular pathways involved, the Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling pathway plays a central role. Dysregulation of the JAK-STAT pathway is implicated in the pathogenesis of CVD by influencing the aforementioned cell types and associated pathological processes. Importantly, the role of the JAK-STAT pathway varies across different types of CVD and throughout different stages of disease progression (e.g., pre-morbid, acute, and chronic phases). This review examines the composition, activation, and regulation of the JAK-STAT pathway and summarizes recent findings on its involvement in CVD. We discuss the distinct roles of JAK-STAT signaling in various CVD conditions, the potential reasons for these differences, and explore the clinical translational prospects and technical challenges of targeting the JAK-STAT pathway for therapeutic intervention in CVD.
Collapse
Affiliation(s)
- Jia-Wei Wu
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, Jiangsu Province, China
- Wuxi Neurosurgical Institute, Wuxi, 214002, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Bing-Xin Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Li-Ping Shen
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, Jiangsu Province, China
- Wuxi Neurosurgical Institute, Wuxi, 214002, Jiangsu Province, China
| | - Yong-Lin Chen
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, Jiangsu Province, China
- Wuxi Neurosurgical Institute, Wuxi, 214002, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Zhi-Yong Du
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, Jiangsu Province, China
- Wuxi Neurosurgical Institute, Wuxi, 214002, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Shi-Qing Du
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, Jiangsu Province, China
- Wuxi Neurosurgical Institute, Wuxi, 214002, Jiangsu Province, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China
| | - Xiao-Jie Lu
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, Jiangsu Province, China.
- Wuxi Neurosurgical Institute, Wuxi, 214002, Jiangsu Province, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu Province, China.
| | - Xu-Dong Zhao
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, 214002, Jiangsu Province, China.
- Wuxi Neurosurgical Institute, Wuxi, 214002, Jiangsu Province, China.
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
7
|
Zhang C, Zhou T, Qiao S, Lu L, Zhu M, Wang A, Zhang S. Taurine Attenuates Neuronal Ferroptosis by CSF-Derived Exosomes of GABABR Encephalitis Through GABABR/NF2/P-YAP Pathway. Mol Neurobiol 2025:10.1007/s12035-025-04819-3. [PMID: 40085353 DOI: 10.1007/s12035-025-04819-3] [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: 09/22/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
GABAB receptor (GABABR) encephalitis represents a rare subtype of paraneoplastic limbic encephalitis (LE), characterized by persistent seizures and cognitive impairments. Nevertheless, the precise phenotype and underlying mechanisms of neuronal dysfunction associated with intrathecal lymphocytes in GABABR encephalitis remain inadequately understood. In the present study, we demonstrate that exosomes derived from the cerebrospinal fluid (CSF) of patients with GABABR encephalitis can induce neuronal ferroptosis, oxidative stress, iron accumulation, and lipid hyperoxidation in an in vitro model of anti-GABABR encephalitis. MicroRNA (miRNA) sequencing revealed that miR-92a-3p is a differentially expressed miRNA in CSF exosomes, and its expression was positively correlated with unfavorable clinical outcomes in GABABR encephalitis patients during a 6-month follow-up period. The NF2/P-YAP signaling pathway was identified as a downstream effector of miR-92a-3p, influencing the expression of ACSL4/GPX4 and IL-6, with the expression of these genes being enhanced following taurine supplementation. Clinically, taurine levels in CSF exhibited a negative correlation with IL-6 levels, CSF cell counts, blood-CSF barrier integrity, and clinical prognosis in GABABR encephalitis. Mechanistically, taurine effectively reduced reactive oxygen species (ROS) and iron accumulation, as well as IL-6 production, while modulating the levels of NF2, P-YAP, ACSL4, and GPX4 in neurons treated with CSF-derived exosomes from GABABR encephalitis through GABABR activation. Proliferation assays indicated that extracellular taurine intake activated CD4 + T cells, CD8 + T cells, and CD19 + B cells in the CSF of patients with GABABR encephalitis. In summary, our findings reveal for the first time that intrathecal lymphocytes in GABABR encephalitis maintain an activated state by absorbing extracellular taurine and that decreased taurine levels in CSF promote neuronal ferroptosis via the miR-92a-3p-mediated NF2/P-YAP/ACSL4 pathway.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China
- Shandong First Medical University, Jinan, China
| | - Tianyu Zhou
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China
- Shandong First Medical University, Jinan, China
| | - Shan Qiao
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China
| | - Lu Lu
- Department of Neurology, Linyi People's Hospital, Linyi, China
| | - Meirong Zhu
- Department of Critical Care Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Aihua Wang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China
| | - Shanchao Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Institute of Neuroimmunology, Jinan, China.
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
8
|
Xu Y, Liu Y, Wu Y, Sun J, Lu X, Dai K, Zhang Y, Luo C, Zhang J. Curcumin Alleviates Microglia-Mediated Neuroinflammation and Neuronal Ferroptosis Following Experimental Subarachnoid Hemorrhage by Modulating the Nrf2/HO-1 Signaling Pathway. Mol Neurobiol 2025; 62:2995-3010. [PMID: 39207623 DOI: 10.1007/s12035-024-04443-7] [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: 01/20/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Early brain injury caused by subarachnoid hemorrhage (SAH) is associated with inflammatory response and ferroptosis. Curcumin alleviates neuroinflammation and oxidative stress by as yet unknown neuroprotective mechanisms. The objective of this study was to investigate the impact of curcumin on neuronal ferroptosis and microglia-induced neuroinflammation following SAH. By examining Nrf2/HO-1 expression levels and ferroptosis biomarkers expression both in vitro and in vivo, it was demonstrated that curcumin effectively suppressed ferroptosis in neurons after SAH through modulation of the Nrf2/HO-1 signaling pathway. Furthermore, by analyzing the expression levels of Nrf2, HO-1, p-p65, and inflammation-related genes, it was confirmed that curcumin could prevent the upregulation of pro-inflammatory factors following SAH by regulating the Nrf2/HO-1/NF-κB signaling pathway in microglia. The ability of curcumin to reduce neuronal damage and cerebral edemas after SAH in mice was validated using TUNEL staining, Nissl staining, and measurement of brain tissue water content. Additionally, through implementation of the modified Garcia test, open field test, and Y-maze test, it was established that curcumin ameliorated neurobehavioral impairments in mice post-SAH. Taken together, these data suggest that curcumin may offer a promising therapeutic approach for improving outcomes following SAH by concurrently attenuating neuronal ferroptosis and reducing neuroinflammation.
Collapse
Affiliation(s)
- Yao Xu
- Department of Emergency Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- National Regional Center for Trauma Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yongsheng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Wu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingshan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaocheng Lu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kun Dai
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yiting Zhang
- Department of Rheumatology, Suzhou Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Suzhou, China.
| | - Chengliang Luo
- Department of Forensic Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China.
| | - Jian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
9
|
Jiang GY, Yang HR, Li C, Liu N, Ma SJ, Jin BX, Yan C, Gong HD, Li JY, Yan HC, Ye GX, Wang WY, Gao C. Ginsenoside Rd alleviates early brain injury by inhibiting ferroptosis through cGAS/STING/DHODH pathway after subarachnoid hemorrhage. Free Radic Biol Med 2025; 228:299-318. [PMID: 39746578 DOI: 10.1016/j.freeradbiomed.2024.12.058] [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: 11/12/2024] [Revised: 12/25/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Ferroptosis, a recently identified form of regulated cell death, is characterized by lipid peroxidation and iron accumulation, plays a critical role in early brain injury after subarachnoid hemorrhage. Ginsenoside Rd, an active compound isolated from ginseng, is known for its neuroprotective properties. However, its influence on SAH-induced ferroptosis remains unclear. In this study, we constructed an SAH model using intravascular perforation in vivo and treated HT22 cells with oxyhemoglobin to simulate the condition in vitro. We observed significant changes in ferroptosis markers, including GPX4 and ACSL4, following SAH. Administration of ginsenoside Rd to both rats and HT22 cells effectively inhibited neuronal ferroptosis induced by SAH, alleviating neurological deficits and cognitive dysfunction in rats. Notably, the neuroprotective properties of ginsenoside Rd were countered by the STING pathway agonist 2'3'-cGAMP. Experiments conducted in vitro and in vivo illustrated that the impacts of ginsenoside Rd were counteracted by the BQR inhibitor. Our findings suggest that ginsenoside Rd mitigates EBI after SAH by suppressing neuronal ferroptosis through the cGAS/STING pathway while upregulating DHODH levels. These outcomes emphasize the potential of ginsenoside Rd as a therapeutic candidate for subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Guang-You Jiang
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hong-Rui Yang
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chen Li
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Nan Liu
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Sheng-Ji Ma
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing-Xuan Jin
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Cong Yan
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hai-Dong Gong
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ji-Yi Li
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao-Chen Yan
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guang-Xi Ye
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wen-Yu Wang
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Cheng Gao
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China; Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
10
|
Naderi S, Khodagholi F, Janahmadi M, Motamedi F, Torabi A, Batool Z, Heydarabadi MF, Pourbadie HG. Ferroptosis and cognitive impairment: Unraveling the link and potential therapeutic targets. Neuropharmacology 2025; 263:110210. [PMID: 39521042 DOI: 10.1016/j.neuropharm.2024.110210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases, share key characteristics, notably cognitive impairment and significant cell death in specific brain regions. Cognition, a complex mental process allowing individuals to perceive time and place, is disrupted in these conditions. This consistent disruption suggests the possibility of a shared underlying mechanism across all neurodegenerative diseases. One potential common factor is the activation of pathways leading to cell death. Despite significant progress in understanding cell death pathways, no definitive treatments have emerged. This has shifted focus towards less-explored mechanisms like ferroptosis, which holds potential due to its involvement in oxidative stress and iron metabolism. Unlike apoptosis or necrosis, ferroptosis offers a novel therapeutic avenue due to its distinct biochemical and genetic underpinnings, making it a promising target in neurodegenerative disease treatment. Ferroptosis is distinguished from other cellular death mechanisms, by distinctive characteristics such as an imbalance of iron hemostasis, peroxidation of lipids in the plasma membrane, and dysregulated glutathione metabolism. In this review, we discuss the potential role of ferroptosis in cognitive impairment. We then summarize the evidence linking ferroptosis biomarkers to cognitive impairment brought on by neurodegeneration while highlighting recent advancements in our understanding of the molecular and genetic mechanisms behind the condition. Finally, we discuss the prospective therapeutic implications of targeting ferroptosis for the treatment of cognitive abnormalities associated with neurodegeneration, including natural and synthetic substances that suppress ferroptosis via a variety of mechanisms. Promising therapeutic candidates, including antioxidants and iron chelators, are being explored to inhibit ferroptosis and mitigate cognitive decline.
Collapse
Affiliation(s)
- Soudabeh Naderi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahyar Janahmadi
- Neuroscience Research Center, Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolfazl Torabi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | | | - Hamid Gholami Pourbadie
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
11
|
Zhou Y, Li J, Yuan Y, Zhang H, Luo X, Wang F, Tao Y, Yue J, Huang L, Wu L, Cao Y, Yu Q, He Q. Metrnl/C-KIT Axis Attenuates Early Brain Injury Following Subarachnoid Hemorrhage by Inhibiting Neuronal Ferroptosis. CNS Neurosci Ther 2025; 31:e70286. [PMID: 39981761 PMCID: PMC11843251 DOI: 10.1111/cns.70286] [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: 11/05/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND AND PURPOSE Ferroptosis is a distinct form of cell death characterized by iron-dependent lipid peroxidation and plays a crucial role in the early brain injury (EBI) following subarachnoid hemorrhage (SAH). As a newly discovered endogenous ligand for the C-KIT receptor tyrosine kinase, meteorin-like protein (Metrnl) exerts regulatory functions in oxidative stress and protects against various diseases. However, the specific role of the Metrnl/C-KIT axis in neuronal ferroptosis during EBI following SAH remains to be elucidated. METHODS Sprague Dawley rats were used to establish the SAH model through endovascular perforation. r-Metrnl was administered intranasally 1 h after SAH. Metrnl shRNA, C-KIT inhibitor ISCK03, AMPK inhibitor dorsomorphin, and Nrf2 inhibitor ML385 were administered intracerebroventricularly or intraperitoneally before r-Metrnl treatment to explore the underlying mechanisms. Neurobehavioral assessments, immunofluorescence, western blot, ELISA, Fluoro-Jade C staining, transmission electron microscopy, and Nissl staining were conducted to evaluate the effects. Additionally, primary neuron culture with hemoglobin (Hb) stimulation was used for in vitro studies. RESULTS Phosphorylated C-KIT and endogenous Metrnl levels were upregulated after SAH. Knockdown of Metrnl aggravated neurobehavioral deficits and neuronal ferroptosis, whereas r-Metrnl treatment showed a protective effect. Mechanistically, r-Metrnl significantly increased the protein levels of SLC7A11, GPX4, FTH, FSP1, and GSH, whereas it decreased the levels of ACSL4, 4HNE, and MDA in the ipsilateral hemisphere 24 h after SAH. Also, r-Metrnl reduced mitochondrial shrinkage, increased mitochondrial crista, and decreased membrane density. However, the beneficial effects of r-Metrnl were partially reversed by ISCK03, dorsomorphin, or ML385 treatment both in vivo and in vitro. CONCLUSIONS Our study demonstrated that r-Metrnl reduced neuronal ferroptosis and improved neurological outcomes after SAH by modulating the C-KIT/AMPK/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- You Zhou
- Department of Critical Care Medicine, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Jiani Li
- Department of Neurology, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Ye Yuan
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Xu Luo
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Feng Wang
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Yihao Tao
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Jianhe Yue
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Luyi Huang
- Key Laboratory of Molecular Biology for Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Lei Wu
- Department of NeurologyGuangdong Second Provincial General HospitalGuangzhouGuangdongChina
| | - Yunxing Cao
- Department of Critical Care Medicine, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| | - Qian Yu
- Department of Neurosurgery, School of Medicine, The Second Affiliated HospitalZhejiang UniversityHangzhouZhejiangChina
| | - Qiuguang He
- Department of Neurosurgery, The Second Affiliated HospitalChongqing Medical UniversityChongqingChina
| |
Collapse
|
12
|
Yang BSK, Savarraj JP, Chen H, Hinds SN, Torres GL, Ryan AS, Atem FD, Lorenzi PL, Ren XS, Badjatia N, Choi HA, Gusdon AM. Systemic Metabolic Alterations after Aneurysmal Subarachnoid Hemorrhage: A Plasma Metabolomics Approach. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.06.25320083. [PMID: 39830284 PMCID: PMC11741492 DOI: 10.1101/2025.01.06.25320083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background Aneurysmal subarachnoid hemorrhage (aSAH) causes systemic changes that contribute to delayed cerebral ischemia (DCI) and morbidity. Circulating metabolites reflecting underlying pathophysiological mechanisms warrant investigation as biomarker candidates. Methods Blood samples, prospectively collected within 24 hours (T1) of admission and 7-days (T2) post ictus, from patients with acute aSAH from two tertiary care centers were retrospectively analyzed. Samples from healthy subjects and patients with non-neurologic critical illness served as controls. A validated external analysis platform was used to perform untargeted metabolomics. Bioinformatics analyses were conducted to identify metabolomic profiles defining each group and delineate metabolic pathways altered in each group. Machine learning (ML) models were developed incorporating key metabolites to improve DCI prediction. Results Among 70 aSAH, 30 healthy control, and 17 sick control subjects, a total of 1,117 metabolites were detected. Groups were matched among key clinical variables. DCI occurred in 36% of aSAH subjects, and poor functional outcome was observed in 70% at discharge. Metabolomic profiles readily discriminated the groups. aSAH subjects demonstrated a robust mobilization of lipid metabolites, with increased levels of free fatty acids (FFAs), mono- and diacylglycerols (MAG, DAG) compared with both control groups. aSAH subjects also had decreased circulating amino acid derived metabolites, consistent with increased catabolism. DCI was associated with increased sphingolipids (sphingosine and sphinganine) and decreased acylcarnitines and S-adenosylhomocysteine at T1. Decreased lysophospholipids and acylcarnitines were associated with poor outcomes. Incorporating metabolites into ML models improved prediction of DCI compared with clinical variables alone. Conclusions Profound metabolic shifts occur after aSAH with characteristic increases in lipid and decreases in amino acid metabolites. Key lipid metabolites associated with outcomes (sphingolipids, lysophospholipids, and acylcarnitines) provide insight into systemic changes driving secondary complications. These metabolites may also prove to be useful biomarkers to improve prognostication and personalize aSAH care.
Collapse
Affiliation(s)
- Bosco Seong Kyu Yang
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Jude P.J. Savarraj
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Hua Chen
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Sarah N. Hinds
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Glenda L. Torres
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Alice S. Ryan
- Department of Medicine, Division of Gerontology, Geriatric, and Palliative Medicine, Geriatric Research, Education, and Clinical Center (GRECC), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Folefac D. Atem
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center, Houston, TX, USA
| | - Philip L. Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computations Biology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA
| | - Xuefang S. Ren
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Neeraj Badjatia
- Program in Trauma, Shock Trauma Neurocritical Care and Department of Neurology, University of Maryland School of Medicine, Baltimore, USA University of Maryland School of Medicine, Baltimore, MD, USA
| | - Huimahn A. Choi
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Aaron M. Gusdon
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
13
|
Zheng B, Zhou X, Pang L, Che Y, Qi X. Dihydroquercetin Ameliorates Neuronal Ferroptosis in Rats After Subarachnoid Hemorrhage via the PI3K/AKT/Nrf2/HO-1 Pathway. J Biochem Mol Toxicol 2025; 39:e70099. [PMID: 39756058 DOI: 10.1002/jbt.70099] [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: 05/17/2024] [Revised: 10/17/2024] [Accepted: 12/08/2024] [Indexed: 01/07/2025]
Abstract
Subarachnoid hemorrhage (SAH) is a specific type of stroke. Dihydroquercetin (DHQ), a flavonoid, is known for its various pharmacological properties. This study aimed to explore the roles and mechanisms of DHQ in influencing the progression of SAH. A rat SAH model was established using the endovascular perforation technique. Following SAH induction, DHQ was administered orally 1 h later. Assessments included SAH scores, neurological function, brain swelling, blood-brain barrier (BBB) integrity, neuronal damage, apoptosis levels, inflammation, and indicators of ferroptosis using various treatments. The HT22 cells were exposed to hemin to simulate SAH-like conditions under in vitro settings. Cell counting kit-8 assays, flow cytometry, enzyme?linked immunosorbent assay, BODIPY 581/591 C11 staining, western blot analysis, and biochemical kits were employed to evaluate the potential effects of DHQ. Moreover, the mechanisms responsible for the protective effect of DHQ were examined by western blot analysis. The in vivo findings revealed that DHQ mitigated neurological impairments, brain swelling, BBB disruption, and neuronal injury at 24 h post-SAH. DHQ also reduced neuronal degeneration, inflammation, and ferroptosis following SAH. The in vitro findings revealed that DHQ enhanced cell survival and reduced ferroptosis at 24 h following hemin exposure. Mechanistically, DHQ activated phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling in SAH rats and hemin-treated HT22 cells to exert neuroprotective effects. In conclusion, this study reveals that DHQ can effectively decrease BBB permeability, brain edema, neurological dysfunctions, and ferroptosis post-SAH by activating the PI3K/AKT/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Bao Zheng
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| | - Xiwei Zhou
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| | - Lujun Pang
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| | - Yanjun Che
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| | - Xin Qi
- Department of Neurosurgery, Jingjiang People's Hospital, Jingjiang, China
| |
Collapse
|
14
|
Hu Y, Gu J, Jin X, Wu X, Li H, Bai L, Wu J, Li X. Asiatic acid alleviates subarachnoid hemorrhage-induced brain injury in rats by inhibiting ferroptosis of neurons via targeting acyl-coenzyme a oxidase 1. Neuropharmacology 2025; 262:110208. [PMID: 39500389 DOI: 10.1016/j.neuropharm.2024.110208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/21/2024] [Accepted: 11/02/2024] [Indexed: 11/11/2024]
Abstract
The occurrence of subarachnoid hemorrhage (SAH) can lead to brain injury, which is a fatal condition with limited effective clinical intervention strategies. The naturally occurring component Asiatic acid (AA), found in the tropical plant Centella asiatica, has been reported to possess neuroprotective properties. The objective of this study was to evaluate the neuroprotective effect of AA following SAH and investigate its potential mechanisms. The SAH model was established in male Sprague-Dawley (SD) rats through intravascular perforation, following a standardized protocol. The administration of AA was performed via gavage following SAH. A lentiviral vector was constructed and utilized for the knockdown of Acyl Coenzyme A Oxidase 1 (ACOX1) Firstly, AA treatment effectively improves brain neurological deficit, neuronal damage, and iron deposition induced by SAH. Furthermore, it has been demonstrated that AA directly interacts with ACOX1, which exhibits decreased expression in neurons following SAH. Additionally, our study reveals AA can reverse SAH-induced reduction in ACOX1 expression, concurrently ameliorating neuronal ferroptosis. This improvement is evidenced by reduced lipid peroxidation, including mitigated GSH depletion, decreased MDA production, and increased GPX4 content and activity. Also, AA enhances mitochondrial constriction while alleviating cristae disruption induced by SAH, providing crucial insights into its neuroprotective effects against neuronal ferroptosis in SAH. Moreover, when ACOX1 is knocked down, the neuroprotective effects of AA are weakened. Collectively, this study elucidated the neuroprotective effect of AA by inhibiting neuronal cell ferroptosis through targeting ACOX1. These findings suggest that AA holds promise as a potential therapeutic candidate for ameliorating SAH-induced brain injury.
Collapse
Affiliation(s)
- Yukun Hu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China; Affliated hospital of Nantong University, Changshu, 215500, Suzhou, Jiangsu Province, China
| | - Jingyu Gu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Xin Jin
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Xiaoxiao Wu
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, 215002, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Lei Bai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| |
Collapse
|
15
|
Ge S, Jing Z, Wang L, Cui X, Zhang X, Wang X. Iron Metabolism and Ferroptosis in Early Brain Injury after Subarachnoid Haemorrhage. Mol Neurobiol 2024; 61:10736-10746. [PMID: 38777982 PMCID: PMC11584420 DOI: 10.1007/s12035-024-04218-0] [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: 07/04/2023] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
At present, it appears that the prognosis for subarachnoid haemorrhage (SAH), which has a high death and disability rate, cannot be greatly improved by medication or other treatment. Recent research suggests that different types of cell death are implicated in early brain injury (EBI) after SAH, and this has been recognised as a major factor impacting the prognosis of SAH. Ferroptosis, which is a recently identified imbalance of iron metabolism and programmed cell death triggered by phospholipid peroxidation, has been shown to be involved in EBI after SAH and is thought to have a significant impact on EBI. The decomposition of cleaved haemoglobin during SAH involves the release of enormous amounts of free iron, resulting in iron metabolism disorders. Potential therapeutic targets for the signalling pathways of iron metabolism disorders and ferroptosis after SAH are constantly being discovered. To serve as a guide for research into other possible therapeutic targets, this paper will briefly describe the mechanisms of dysregulated iron metabolism and ferroptosis in the pathogenesis of SAH and highlight how they are involved in the development and promotion of EBI in SAH.
Collapse
Affiliation(s)
- Shihao Ge
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Ziwen Jing
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Lele Wang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xiaocong Cui
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xin Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xiaopeng Wang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
16
|
Yang J, Wu Q, Li Y, Zhang Y, Lan S, Yuan K, Dai J, Sun B, Meng Y, Xu S, Shi H. BL-918 alleviates oxidative stress in rats after subarachnoid hemorrhage by promoting mitophagy through the ULK1/PINK1/Parkin pathway. Free Radic Biol Med 2024; 224:846-861. [PMID: 39368518 DOI: 10.1016/j.freeradbiomed.2024.10.261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND AND PURPOSE Oxidative stress plays a critical role in early brain injury (EBI) following subarachnoid hemorrhage (SAH). The small molecule ULK1 agonist, BL-918, demonstrated neuroprotective effects in other central nervous system diseases; however, its role in SAH has not yet been explored. This study aimed to evaluate whether BL-918 could provide neuroprotective effects in rats following SAH. METHODS An SAH model was established in Sprague-Dawley rats using endovascular perforation. BL-918 was administered intraperitoneally after SAH, while the ULK1 inhibitor SBI was given intraperitoneally prior to SAH modeling. PINK1 siRNA was administered into the lateral ventricle before SAH induction. The neuroprotective effects and mechanisms of BL-918 were assessed through SAH grading, brain water content measurement, blood-brain barrier permeability, neurobehavioral tests, Western blot, immunofluorescence, TUNEL staining, DHE staining, and transmission electron microscopy (TEM). RESULTS After SAH, the expression levels of p-ULK1, PINK1, Parkin, and LC3Ⅱ increased, peaking at 24 h post-SAH. BL-918 treatment improved neurological function in rats, reduced brain water content and blood-brain barrier permeability, and exhibited anti-oxidative stress and anti-apoptotic effects. Western blot analysis revealed that BL-918 increased the expression of p-ULK1, PINK1, Parkin, LC3Ⅱ, Bcl-xl, and Bcl-2 while inhibiting the expression of Bax and Cleaved Caspase-3. Oxidative stress-related indicators showed that BL-918 alleviated oxidative stress. Immunofluorescence and TEM results demonstrated that BL-918 promoted mitophagy and preserved mitochondrial morphology. Furthermore, the positive effects of BL-918 were reversed by SBI and PINK1 siRNA, respectively. CONCLUSION BL-918 improved both short-term and long-term neurological impairments in rats after SAH and reduced oxidative stress by promoting mitophagy, at least partially through the ULK1/PINK1/Parkin signaling pathway.
Collapse
Affiliation(s)
- Jinshuo Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiaowei Wu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuchen Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongzhi Zhang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuai Lan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kaikun Yuan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaxing Dai
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bowen Sun
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuxiao Meng
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shancai Xu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Huaizhang Shi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
17
|
Zhao Y, Guo H, Li Q, Wang N, Yan C, Zhang S, Dong Y, Liu C, Gao W, Zhu Y, Li Q. TREM1 induces microglial ferroptosis through the PERK pathway in diabetic-associated cognitive impairment. Exp Neurol 2024; 383:115031. [PMID: 39461708 DOI: 10.1016/j.expneurol.2024.115031] [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: 07/11/2024] [Revised: 09/28/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Ferroptosis is involved in neurodegenerative disorders including diabetes-associated cognitive impairment (DACI). As central immune cells, microglia have strong siderophilic properties. However, the role of iron deposition in microglia and the underlying regulatory mechanism remains unclear in DACI. Here, we established high glucose (HG) model in BV2/HMC3 cells and diabetes model in C57BL/6 J mice with HFD and STZ. Transmission Electron Microscopy, Western blot, assay kits of Fe2+, GSH/GSSG, MDA and ROS were carried out in vitro. Prussian blue staining, Western blot and immunofluorescence were implemented in vivo. Y-maze and novel object recognition were performed to assess cognitive performance. LP17 was used to inhibit TREM1 (triggering receptor expressed on myeloid cells 1) specifically in vivo and vitro. We found excessively deposited iron and significant reduction in antioxidants in hippocampal microglia of mice with DACI, concomitant with increased TREM1 (a microglia-specific inflammatory amplifier). Furthermore, LP17 (TREM1 specific inhibitor) ameliorated cognitive impairment caused by HFD/STZ through relieving iron accumulation and antioxidant inactivation. In vitro, ferroptosis was induced by HG in mice microglia-BV2 and human microglia-HMC3 cells, which could be blocked by a ferroptosis inhibitor-Fer-1 and LP17. Moreover, PERK pathway of endoplasmic reticulum stress was activated by HG, and then reversed by PERK inhibitor GSK2606414 and LP17 followed by improved ferroptosis in HG-cultured BV2. In summary, our results indicated that TREM1 effectively aggravates T2DM-associated microglial iron accumulation through the PERK pathway of ERS, which contributes to antioxidant inactivation and lipid peroxidation, eventually, massively boosted ROS result in microglial ferroptosis. The mechanism elucidation in our study may shed light on targeted therapy of DACI.
Collapse
Affiliation(s)
- Yujing Zhao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Hongyan Guo
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Qiao Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China; Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Nan Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Chaoying Yan
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Simei Zhang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Yicong Dong
- Center of Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Chang Liu
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Wei Gao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Yaomin Zhu
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Qing Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
18
|
Lu J, Xu H, Li L, Tang X, Zhang Y, Zhang D, Xu P, Sun L, Wang J. Didang Tang alleviates neuronal ferroptosis after intracerebral hemorrhage by modulating the PERK/eIF2α/ATF4/CHOP/GPX4 signaling pathway. Front Pharmacol 2024; 15:1472813. [PMID: 39525631 PMCID: PMC11544539 DOI: 10.3389/fphar.2024.1472813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Ferroptosis is a crucial process contributing to neuronal damage following intracerebral hemorrhage (ICH). Didang Tang (DDT), a traditional therapeutic, has been used clinically to manage ICH for many years, yet the molecular mechanisms by which by DDT protects neurons from ferroptosis after ICH remain elusive. Methods This study utilized high-performance liquid chromatography-based fingerprint analysis to characterize DDT's chemical composition. An ICH rat model and hemin and erastin-induced PC12 cell ferroptosis models were developed to investigate DDT's neuroprotective mechanisms. Histological assessments of brain tissue morphology and iron deposition were performed using hematoxylin-eosin, Nissl, and Perl's blue staining. Neurological function was evaluated using Longa and Berderson scores, while lipid peroxidation was measured using biochemical assays and flow cytometry. Protein expression levels of ferroptosis- and endoplasmic reticulum stress (ERS)-related markers were analyzed via Western blotting and immunofluorescence. Results Our results demonstrated that DDT reduced hematoma volume, decreased iron deposition, lowered malondialdehyde (MDA) levels, and upregulated glutathione peroxidase (GPX4) and SLC7A11 expression in affected brain regions. Furthermore, DDT downregulated GRP78 expression and inhibited the PERK/eIF2α/ATF4/CHOP/GPX4 pathway, exerting strong neuroprotective effects. The fluorescence staining results of MAP2/GPX4 and MAP2/CHOP suggested that DDT may regulate neuronal ferroptosis and ERs to exert the protective effect. In vitro experiments using hemin- and erastin-induced neuron-derived PC12 cells as neuronal ferroptosis models developed in our laboratory corroborated these in vivo findings, showing increased survival and reduced lipid peroxidation in DDT-treated cells, along with similar inhibitory effects on ferroptosis and ERS. Molecular docking suggested that DDT components, such as sennoside B, amygdalin, rhein, and emodin, interact favorably with PERK/eIF2α/ATF4/CHOP signaling pathway proteins, highlighting their potential role in DDT's anti-ferroptosis effects. Conclusion DDT alleviates neuronal ferroptosis after ICH by modulating the PERK/eIF2α/ATF4/CHOP/GPX4 signaling pathway. Overall, this study provides novel insights into DDT's protective mechanisms against ICH-induced neuronal injury by modulating ferroptosis and ERS pathways, underscoring its potential as an effective therapeutic strategy.
Collapse
Affiliation(s)
- Jing Lu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Hanying Xu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
- Department of Encephalopathy, The Affiliated Hospital of Changchun University of Chinese Medicine, Jilin, China
| | - Li Li
- Nursing Department, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Xiaolei Tang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Ying Zhang
- Department of Encephalopathy, The Affiliated Hospital of Changchun University of Chinese Medicine, Jilin, China
| | - Dongmei Zhang
- Scientific Research Office, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Peng Xu
- Department of Encephalopathy, The Affiliated Hospital of Changchun University of Chinese Medicine, Jilin, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Jian Wang
- Department of Encephalopathy, The Affiliated Hospital of Changchun University of Chinese Medicine, Jilin, China
| |
Collapse
|
19
|
Yang J, Wu Q, Lan S, Yuan K, Sun B, Meng Y, Xu S, Shi H. Peroxiredoxin-5 alleviates early brain injury after subarachnoid hemorrhage by reducing oxidative stress. Brain Res Bull 2024; 217:111087. [PMID: 39326715 DOI: 10.1016/j.brainresbull.2024.111087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND PURPOSE Following subarachnoid hemorrhage (SAH), excessive activation of oxidative stress and cell apoptosis plays a critical role in early brain injury (EBI). Peroxiredoxin-5 (Prdx5), predominantly expressed in neuronal mitochondria, acts as an antioxidant. However, the role of Prdx5 in EBI after SAH remains unclear. This study aims to elucidate the antioxidative stress and anti-apoptotic effects of Prdx5 in rats following SAH. METHODS In this study, an SAH model was established in Sprague-Dawley rats using endovascular perforation. Recombinant Prdx5 (rPrdx5) was administered intranasally to upregulate Prdx5 expression after SAH in rats. Prdx5 small interfering RNA (Prdx5 siRNA) was administered prior to SAH modelling. The neuroprotective effects of Prdx5 were validated through SAH grading, brain water content, blood-brain barrier permeability, neurobehavioral tests, immunofluorescence, TUNEL staining, and Western blotting. RESULTS The expression levels of endogenous Prdx5 significantly decreased after SAH. Treatment with rPrdx5 improved both short-term and long-term behaviour in rats, reduced brain water content and blood-brain barrier permeability, and exhibited anti-oxidative stress and anti-apoptotic effects. Measurements of oxidative stress-related indicators, including MDA, SOD, GSH-Px and GSH/GSSG, confirmed that Prdx5 can alleviate oxidative stress in rats after SAH. Western blot analysis showed that rPrdx5 significantly increased the expression of Bcl-XL and Bcl-2 and reduced the expression of Bax and Cleaved Caspase-3, thereby exerting anti-apoptotic effects. Additionally, Prdx5 siRNA reversed the neuroprotective effects of rPrdx5, exacerbated neuronal damage and blood-brain barrier permeability, and increased levels of oxidative stress and apoptosis. CONCLUSION In conclusion, our study demonstrated that specifically upregulating the expression of Prdx5 can reduce oxidative stress and apoptosis in rats after SAH, while also improving both short-term and long-term neurological impairments. Prdx5 is primarily expressed in the mitochondria of neuronal cells and is a crucial target for reducing ROS after SAH. rPrdx5 treatment may offer a promising therapeutic approach for clinical SAH patients.
Collapse
Affiliation(s)
- Jinshuo Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiaowei Wu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuai Lan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kaikun Yuan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bowen Sun
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuxiao Meng
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shancai Xu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Huaizhang Shi
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
20
|
Xiao F, Li HL, Yang B, Che H, Xu F, Li G, Zhou CH, Wang S. Disulfidptosis: A new type of cell death. Apoptosis 2024; 29:1309-1329. [PMID: 38886311 PMCID: PMC11416406 DOI: 10.1007/s10495-024-01989-8] [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: 05/28/2024] [Indexed: 06/20/2024]
Abstract
Disulfidptosis is a novel form of cell death that is distinguishable from established programmed cell death pathways such as apoptosis, pyroptosis, autophagy, ferroptosis, and oxeiptosis. This process is characterized by the rapid depletion of nicotinamide adenine dinucleotide phosphate (NADPH) in cells and high expression of solute carrier family 7 member 11 (SLC7A11) during glucose starvation, resulting in abnormal cystine accumulation, which subsequently induces andabnormal disulfide bond formation in actin cytoskeleton proteins, culminating in actin network collapse and disulfidptosis. This review aimed to summarize the underlying mechanisms, influencing factors, comparisons with traditional cell death pathways, associations with related diseases, application prospects, and future research directions related to disulfidptosis.
Collapse
Affiliation(s)
- Fei Xiao
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hui-Li Li
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Emergency, The State Key Laboratory for Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Bei Yang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hao Che
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Fei Xu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Gang Li
- Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Cheng-Hui Zhou
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Linzhi People's Hospital, Linzhi, Tibet, China.
| |
Collapse
|
21
|
Liu N, Li C, Yan C, Yan HC, Jin BX, Yang HR, Jiang GY, Gong HD, Li JY, Ma SJ, Liu HL, Gao C. BCAT1 alleviates early brain injury by inhibiting ferroptosis through PI3K/AKT/mTOR/GPX4 pathway after subarachnoid hemorrhage. Free Radic Biol Med 2024; 222:173-186. [PMID: 38871197 DOI: 10.1016/j.freeradbiomed.2024.05.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/22/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Regulation of the redox system by branched-chain amino acid transferase 1 (BCAT1) is of great significance in the occurrence and development of diseases, but the relationship between BCAT1 and subarachnoid hemorrhage (SAH) is still unknown. Ferroptosis, featured by iron-dependent lipid peroxidation accompanied by the depletion of glutathione peroxidase 4 (GPX4), has been implicated in the pathological process of early brain injury after subarachnoid hemorrhage. This study established SAH model by endovascular perforation and adding oxyhemoglobin (Hb) to HT22 cells and delved into the mechanism of BCAT1 in SAH-induced ferroptotic neuronal cell death. It was found that SAH-induced neuronal ferroptosis could be inhibited by BCAT1 overexpression (OE) in rats and HT22 cells, and BCAT1 OE alleviated neurological deficits and cognitive dysfunction in rats after SAH. In addition, the effect of BCAT1 could be reversed by the Ly294002, a specific inhibitor of the PI3K pathway. In summary, our present study indicated that BCAT1 OE alleviated early brain injury EBI after SAH by inhibiting neuron ferroptosis via activation of PI3K/AKT/mTOR pathway and the elevation of GPX4. These results suggested that BCAT1 was a promising therapeutic target for subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Nan Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Chen Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Cong Yan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hao-Chen Yan
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Bing-Xuan Jin
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hong-Rui Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Guang-You Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Hai-Dong Gong
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Ji-Yi Li
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Sheng-Ji Ma
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Huai-Lei Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China.
| | - Cheng Gao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, China; Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China.
| |
Collapse
|
22
|
Song Y, Luo X, Yao L, Chen Y, Mao X. A Novel Mechanism Linking Melatonin, Ferroptosis and Microglia Polarization via the Circodz3/HuR Axis in Subarachnoid Hemorrhage. Neurochem Res 2024; 49:2556-2572. [PMID: 38888828 DOI: 10.1007/s11064-024-04193-x] [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: 03/15/2024] [Revised: 05/23/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
A subarachnoid hemorrhage (SAH) is life-threatening bleeding into the subarachnoid space that causes brain damage. Growing evidence has suggested that melatonin provides neuroprotection following SAH. Exploring the mechanisms underlying melatonin-mediated neuroprotection contributes to its clinical application in SAH. The plasma and cerebrospinal fluid (CSF) were collected from SAH patients, and SAH mice were established via pre-chiasmatic injection. Circodz3 expression, levels of IL-1β and TNF-α, brain water content, neurological and beam-waling scores were determined. Ferroptosis was evaluated by analyzing levels of iron, lipid ROS, MDA, and GSH. The colocalization of circodz3 and Iba-1 was analyzed by immunofluorescence staining. Interaction of circodz3 and HuR was determined with RNA pull-down and RNA immunoprecipitation assays. Herein, we found that circodz3 was highly abundant in SAH patients and mice. Colocalization of circodz3 and Iba-1 in the left hemisphere of SAH mice suggested the implication of circodz3 in regulating microglia activation following SAH. Melatonin alleviated brain edema, neurological impairment, and microglia activation and inhibited circodz3 expression in SAH mice. Moreover, melatonin inhibited M1 polarization, oxidative stress and ferroptosis and restrained circodz3 expression in primary microglia following SAH. These effects were abrogated by circodz3 overexpression. Circodz3 knockdown inhibited ferroptosis and M1 polarization of BV2 microglia after SAH. Circodz3 interacted with HuR to facilitate β-Trcp1-mediated ubiquitination and degradation, thus restraining the expression of SLC7A11 and GPX4. Collectively, melatonin exerted neuroprotection following SAH via inhibiting ferroptosis and M1 polarization through the circodz3/HuR axis. Our study suggests potential application of melatonin in the treatment of SAH.
Collapse
Affiliation(s)
- Yanju Song
- Department of Neurology, The Third Hospital of Changsha, No.176 Laodong West Road, Tianxin District, Changsha, 410015, Hunan Province, People's Republic of China
| | - Xin Luo
- Department of Neurology, The Third Hospital of Changsha, No.176 Laodong West Road, Tianxin District, Changsha, 410015, Hunan Province, People's Republic of China
| | - Liping Yao
- Department of Neurology, The Third Hospital of Changsha, No.176 Laodong West Road, Tianxin District, Changsha, 410015, Hunan Province, People's Republic of China
| | - YingChao Chen
- Department of Neurology, The Third Hospital of Changsha, No.176 Laodong West Road, Tianxin District, Changsha, 410015, Hunan Province, People's Republic of China
| | - Xinfa Mao
- Department of Neurology, The Third Hospital of Changsha, No.176 Laodong West Road, Tianxin District, Changsha, 410015, Hunan Province, People's Republic of China.
| |
Collapse
|
23
|
Zhu W, Dong J, Han Y. Electroacupuncture Downregulating Neuronal Ferroptosis in MCAO/R Rats by Activating Nrf2/SLC7A11/GPX4 Axis. Neurochem Res 2024; 49:2105-2119. [PMID: 38819696 PMCID: PMC11233380 DOI: 10.1007/s11064-024-04185-x] [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: 09/13/2023] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Ischemic stroke involves various pathological processes, among which ferroptosis is crucial. Previous studies by our group have indicated that electroacupuncture (EA) mitigates ferroptosis after ischemic stroke; however, the precise mechanism underlying this effect remains unclear. In the present study, we developed a rat model of middle cerebral artery occlusion/reperfusion. We chose the main acupoint of the treatment methods of the "Awakening and Opening of the Brain". Rats' neurological function and motor coordination were evaluated by neurological function score and the rotarod test, respectively, and the volume of cerebral infarction was analyzed by 2,3,5-triphenyltetrazolium chloride Staining. The cerebrovascular conditions were visualized by time-of-flight magentic resonance angiography. In addition, we detected changes in lipid peroxidation and endogenous antioxidant activity by measuring the malondialdehyde, glutathione, superoxide dismutase activities, glutathione/oxidized glutathione and reduced nicotinamide adenine dinucleotide phosphate/oxidized nicotinamide adenine dinucleotide phosphate ratios. Inductively coupled plasma-mass spectrometry, western blot, reverse transcription-polymerase chain reaction, fluoro-jade B staining, immunofluorescence analysis, and transmission electron microscopy were utilized to examine the influence of EA. The results indicate that EA treatment was effective in reversing neurological impairment, neuronal damage, and protecting mitochondrial morphology and decreasing the cerebral infarct volume in the middle cerebral artery occlusion/reperfusion rat model. EA reduced iron levels, inhibited lipid peroxidation, increased endogenous antioxidant activity, modulated the expression of several ferroptosis-related proteins, and promoted nuclear factor-E2-related factor 2 (Nrf2) nuclear translocation. However, the protective effect of EA was hindered by the Nrf2 inhibitor ML385. These findings suggest that EA can suppress ferroptosis and decrease damage caused by cerebral ischemia/reperfusion by activating Nrf2 and increasing the protein expression of solute carrier family 7 member 11 and glutathione peroxidase 4.
Collapse
Affiliation(s)
- Wei Zhu
- Institute of Neurology, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jianjian Dong
- Institute of Neurology, Anhui University of Chinese Medicine, Hefei, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yongsheng Han
- Institute of Neurology, Anhui University of Chinese Medicine, Hefei, Anhui, China.
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei, Anhui, China.
- Wannan Medical College, Wuhu, Anhui, China.
| |
Collapse
|
24
|
Cong J, Li JY, Zou W. Mechanism and treatment of intracerebral hemorrhage focus on mitochondrial permeability transition pore. Front Mol Neurosci 2024; 17:1423132. [PMID: 39156127 PMCID: PMC11328408 DOI: 10.3389/fnmol.2024.1423132] [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: 04/25/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is the second most common subtype of stroke, characterized by high mortality and a poor prognosis. Despite various treatment methods, there has been limited improvement in the prognosis of ICH over the past decades. Therefore, it is imperative to identify a feasible treatment strategy for ICH. Mitochondria are organelles present in most eukaryotic cells and serve as the primary sites for aerobic respiration and energy production. Under unfavorable cellular conditions, mitochondria can induce changes in permeability through the opening of the mitochondrial permeability transition pore (mPTP), ultimately leading to mitochondrial dysfunction and contributing to various diseases. Recent studies have demonstrated that mPTP plays a role in the pathological processes associated with several neurodegenerative diseases including Parkinson's disease, Alzheimer's disease, Huntington's disease, ischemic stroke and ischemia-reperfusion injury, among others. However, there is limited research on mPTP involvement specifically in ICH. Therefore, this study comprehensively examines the pathological processes associated with mPTP in terms of oxidative stress, apoptosis, necrosis, autophagy, ferroptosis, and other related mechanisms to elucidate the potential mechanism underlying mPTP involvement in ICH. This research aims to provide novel insights for the treatment of secondary injury after ICH.
Collapse
Affiliation(s)
- Jing Cong
- The First School of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing-Yi Li
- The Second School of Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Zou
- Molecular Biology Laboratory of Clinical Integrated of Traditional Chinese and Western Medicine of Heilong Jiang Province, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
25
|
Wu Q, Yuan K, Yao Y, Yao J, Shao J, Meng Y, Wu P, Shi H. LAMC1 attenuates neuronal apoptosis via FAK/PI3K/AKT signaling pathway after subarachnoid hemorrhage. Exp Neurol 2024; 376:114776. [PMID: 38609046 DOI: 10.1016/j.expneurol.2024.114776] [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: 01/17/2024] [Revised: 03/20/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND AND PURPOSE The poor prognosis in patients with subarachnoid hemorrhage (SAH) is often attributed to neuronal apoptosis. Recent evidence suggests that Laminin subunit gamma 1 (LAMC1) is essential for cell survival and proliferation. However, the effects of LAMC1 on early brain injury after SAH and the underlying mechanisms are unknown. The current study aimed to reveal the anti-neuronal apoptotic effect and the potential mechanism of LAMC1 in the rat and in the in vitro SAH models. METHODS The SAH model of Sprague-Dawley rats was established by endovascular perforation. Recombinant LAMC1 (rLAMC1) was administered intranasally 30 min after modeling. LAMC1 small interfering RNA (LAMC1 siRNA), focal adhesion kinase (FAK)-specific inhibitor Y15 and PI3K-specific inhibitor LY294002 were administered before SAH modeling to explore the neuroprotection mechanism of rLAMC1. HT22 cells were cultured and stimulated by oxyhemoglobin to establish an in vitro model of SAH. Subsequently, SAH grades, neurobehavioral tests, brain water content, blood-brain barrier permeability, western blotting, immunofluorescence, TUNEL, and Fluoro-Jade C staining were performed. RESULTS The expression of endogenous LAMC1 was markedly decreased after SAH, both in vitro and in vivo. rLAMC1 significantly reduced the brain water content and blood-brain barrier permeability, improved short- and long-term neurobehavior, and decreased neuronal apoptosis. Furthermore, rLAMC1 treatment significantly increased the expression of p-FAK, p-PI3K, p-AKT, Bcl-XL, and Bcl-2 and decreased the expression of Bax and cleaved caspase -3. Conversely, knockdown of endogenous LAMC1 aggravated the neurological impairment, suppressed the expression of Bcl-XL and Bcl-2, and upregulated the expression of Bax and cleaved caspase-3. Additionally, the administration of Y15 and LY294002 abolished the protective roles of rLAMC1. In vitro, rLAMC1 significantly reduced neuronal apoptosis, and the protective effects were also abolished by Y15 and LY294002. CONCLUSION Exogenous LAMC1 treatment improved neurological deficits after SAH in rats, and attenuated neuronal apoptosis in both in vitro and in vivo SAH models, at least partially through the FAK/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Qiaowei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Kaikun Yuan
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yanting Yao
- Department of Neurosurgery, Beidahuang Group General Hospital, Harbin, Heilongjiang, China
| | - Jinbiao Yao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiang Shao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yuxiao Meng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Pei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
26
|
Liu C, Wang G, Han W, Tian Q, Li M. Ferroptosis: a potential therapeutic target for stroke. Neural Regen Res 2024; 19:988-997. [PMID: 37862200 PMCID: PMC10749612 DOI: 10.4103/1673-5374.385284] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/05/2023] [Accepted: 08/03/2023] [Indexed: 10/22/2023] Open
Abstract
Ferroptosis is a form of regulated cell death characterized by massive iron accumulation and iron-dependent lipid peroxidation, differing from apoptosis, necroptosis, and autophagy in several aspects. Ferroptosis is regarded as a critical mechanism of a series of pathophysiological reactions after stroke because of iron overload caused by hemoglobin degradation and iron metabolism imbalance. In this review, we discuss ferroptosis-related metabolisms, important molecules directly or indirectly targeting iron metabolism and lipid peroxidation, and transcriptional regulation of ferroptosis, revealing the role of ferroptosis in the progression of stroke. We present updated progress in the intervention of ferroptosis as therapeutic strategies for stroke in vivo and in vitro and summarize the effects of ferroptosis inhibitors on stroke. Our review facilitates further understanding of ferroptosis pathogenesis in stroke, proposes new targets for the treatment of stroke, and suggests that more efforts should be made to investigate the mechanism of ferroptosis in stroke.
Collapse
Affiliation(s)
- Chengli Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Guijun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Wenrui Han
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
27
|
Peng Z, Ding YN, Yang ZM, Li XJ, Zhuang Z, Lu Y, Tang QS, Hang CH, Li W. Neuron-targeted liposomal coenzyme Q10 attenuates neuronal ferroptosis after subarachnoid hemorrhage by activating the ferroptosis suppressor protein 1/coenzyme Q10 system. Acta Biomater 2024; 179:325-339. [PMID: 38561074 DOI: 10.1016/j.actbio.2024.03.023] [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: 12/18/2023] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024]
Abstract
Subarachnoid hemorrhage (SAH) is primarily attributed to the rupture of intracranial aneurysms and is associated with a high incidence of disability and mortality. SAH disrupts the blood‒brain barrier, leading to the release of iron ions from blood within the subarachnoid space, subsequently inducing neuronal ferroptosis. A recently discovered protein, known as ferroptosis suppressor protein 1 (FSP1), exerts anti-ferroptotic effects by facilitating the conversion of oxidative coenzyme Q 10 (CoQ10) to its reduced form, which effectively scavenges reactive oxygen radicals and mitigates iron-induced ferroptosis. In our investigation, we observed an increase in FSP1 levels following SAH. However, the depletion of CoQ10 caused by SAH hindered the biological function of FSP1. Therefore, we created neuron-targeted liposomal CoQ10 by introducing the neuron-targeting peptide Tet1 onto the surface of liposomal CoQ10. Our objective was to determine whether this formulation could activate the FSP1 system and subsequently inhibit neuronal ferroptosis. Our findings revealed that neuron-targeted liposomal CoQ10 effectively localized to neurons at the lesion site after SAH. Furthermore, it facilitated the upregulation of FSP1, reduced the accumulation of malondialdehyde and reactive oxygen species, inhibited neuronal ferroptosis, and exerted neuroprotective effects both in vitro and in vivo. Our study provides evidence that supplementation with CoQ10 can effectively activate the FSP1 system. Additionally, we developed a neuron-targeted liposomal CoQ10 formulation that can be selectively delivered to neurons at the site of SAH. This innovative approach represents a promising therapeutic strategy for neuronal ferroptosis following SAH. STATEMENT OF SIGNIFICANCE: Subarachnoid hemorrhage (SAH) is primarily attributed to the rupture of intracranial aneurysms and is associated with a high incidence of disability and mortality. Ferroptosis suppressor protein 1 (FSP1), exerts anti-ferroptotic effects by facilitating the conversion of oxidative coenzyme Q 10 (CoQ10) to its reduced form, which effectively scavenges reactive oxygen radicals and mitigates iron-induced ferroptosis. In our investigation, we observed an increase in FSP1 levels following SAH. However, the depletion of CoQ10 caused by SAH hindered the biological function of FSP1. Therefore, we created neuron-targeted liposomal CoQ10. We find that it effectively localized to neurons at the lesion site after SAH and activated the FSP1/CoQ10 system. This innovative approach represents a promising therapeutic strategy for neuronal ferroptosis following SAH and other central nervous system diseases characterized by disruption of the blood-brain barrier.
Collapse
Affiliation(s)
- Zheng Peng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Yi-Nan Ding
- Department of Interventional Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China.
| | | | - Xiao-Jian Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Zong Zhuang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Yue Lu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Qiu-Sha Tang
- Medical School of Southeast University, Nanjing, China.
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Neurosurgical Institute, Nanjing University, Nanjing, China.
| |
Collapse
|
28
|
Huang L, Wang X, Zheng Y, Lang D, Wang J, Yan S, Chen Y. EGCG-NPs inhibition HO-1-mediated reprogram iron metabolism against ferroptosis after subarachnoid hemorrhage. Redox Biol 2024; 70:103075. [PMID: 38364686 PMCID: PMC10878112 DOI: 10.1016/j.redox.2024.103075] [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: 12/20/2023] [Revised: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 02/18/2024] Open
Abstract
Subarachnoid hemorrhage (SAH), a devastating disease with a high mortality rate and poor outcomes, tightly associated with the dysregulation of iron metabolism and ferroptosis. (-)-Epigallocatechin-3-gallate (EGCG) is one of major bioactive compounds of tea catechin because of its well-known iron-chelating and antioxidative activities. However, the findings of iron-induced cell injuries after SAH remain controversial and the underlying therapeutic mechanisms of EGCG in ferroptosis is limited. Here, the ability of EGCG to inhibit iron-induced cell death following the alleviation of neurological function deficits was investigated by using in vivo SAH models. As expected, EGCG inhibited oxyhemoglobin (OxyHb)-induced the over-expression of HO-1, which mainly distributed in astrocytes and microglial cells. Subsequently, EGCG blocked ferrous iron accumulation through HO-1-mediated iron metabolic reprogramming. Therefore, oxidative stress and mitochondrial dysfunction was rescued by EGCG, which resulted in the downregulation of ferroptosis and ferritinophagy rather than apoptosis after SAH. As a result, EGCG exerted the superior therapeutic effects in the maintenance of iron homeostasis in glial cells, such as astrocytes and microglial cells, as well as in the improvement of functional outcomes after SAH. These findings highlighted that glial cells were not only the iron-rich cells in the brain but also susceptible to ferroptosis and ferritinophagy after SAH. The detrimental role of HO-1-mediated ferroptosis in glial cells can be regarded as an effective therapeutic target of EGCG in the prevention and treatment of SAH.
Collapse
Affiliation(s)
- Liyong Huang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Henan, China; Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Henan, China
| | - Xue Wang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Yanning Zheng
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Dongcen Lang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Jian Wang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Shuaiguo Yan
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Ying Chen
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China.
| |
Collapse
|
29
|
Li S, Huang P, Lai F, Zhang T, Guan J, Wan H, He Y. Mechanisms of Ferritinophagy and Ferroptosis in Diseases. Mol Neurobiol 2024; 61:1605-1626. [PMID: 37736794 DOI: 10.1007/s12035-023-03640-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
The discovery of the role of autophagy, particularly the selective form like ferritinophagy, in promoting cells to undergo ferroptosis has inspired us to investigate functional connections between diseases and cell death. Ferroptosis is a novel model of procedural cell death characterized by the accumulation of iron-dependent reactive oxygen species (ROS), mitochondrial dysfunction, and neuroinflammatory response. Based on ferroptosis, the study of ferritinophagy is particularly important. In recent years, extensive research has elucidated the role of ferroptosis and ferritinophagy in neurological diseases and anemia, suggesting their potential as therapeutic targets. Besides, the global emergence and rapid transmission of COVID-19, which is caused by SARS-CoV-2, represents a considerable risk to public health worldwide. The potential involvement of ferroptosis in the pathophysiology of brain injury associated with COVID-19 is still unclear. This review summarizes the pathophysiological changes of ferroptosis and ferritinophagy in neurological diseases, anemia, and COVID-19, and hypothesizes that ferritinophagy may be a potential mechanism of ferroptosis. Advancements in these fields will enhance our comprehension of methods to prevent and address neurological disorders, anemia, and COVID-19.
Collapse
Affiliation(s)
- Siqi Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ping Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Feifan Lai
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Ting Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jiaqi Guan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Haitong Wan
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
30
|
Kang J, Tian S, Zhang L, Yang G. Ferroptosis in early brain injury after subarachnoid hemorrhage: review of literature. Chin Neurosurg J 2024; 10:6. [PMID: 38347652 PMCID: PMC10863120 DOI: 10.1186/s41016-024-00357-4] [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: 10/18/2023] [Accepted: 01/28/2024] [Indexed: 02/15/2024] Open
Abstract
Spontaneous subarachnoid hemorrhage (SAH), mainly caused by ruptured intracranial aneurysms, is a serious acute cerebrovascular disease. Early brain injury (EBI) is all brain injury occurring within 72 h after SAH, mainly including increased intracranial pressure, decreased cerebral blood flow, disruption of the blood-brain barrier, brain edema, oxidative stress, and neuroinflammation. It activates cell death pathways, leading to neuronal and glial cell death, and is significantly associated with poor prognosis. Ferroptosis is characterized by iron-dependent accumulation of lipid peroxides and is involved in the process of neuron and glial cell death in early brain injury. This paper reviews the research progress of ferroptosis in early brain injury after subarachnoid hemorrhage and provides new ideas for future research.
Collapse
Affiliation(s)
- Junlin Kang
- The First Hospital of Lanzhou University, Lanzhou City, Gansu Province, China
| | - Shilai Tian
- The First Hospital of Lanzhou University, Lanzhou City, Gansu Province, China
| | - Lei Zhang
- Gansu Provincial Hospital, Lanzhou City, Gansu Province, China
| | - Gang Yang
- The First Hospital of Lanzhou University, Lanzhou City, Gansu Province, China.
| |
Collapse
|
31
|
Wang S, Xiong B, Tian Y, Hu Q, Jiang X, Zhang J, Chen L, Wang R, Li M, Zhou X, Zhang T, Ge H, Yu A. Targeting Ferroptosis Promotes Functional Recovery by Mitigating White Matter Injury Following Acute Carbon Monoxide Poisoning. Mol Neurobiol 2024; 61:1157-1174. [PMID: 37697220 DOI: 10.1007/s12035-023-03603-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023]
Abstract
Survivors experiencing acute carbon monoxide poisoning (ACMP) tend to develop white matter injury (WMI). The mechanism of ACMP-induced WMI remains unclear. Considering the role of ferroptosis in initiating oligodendrocyte damage to deteriorate WMI, exploring therapeutic options to attenuate ferroptosis is a feasible approach to alleviating WMI. Our results indicated that ACMP induced accumulation of iron and reactive oxygen species (ROS) eventually leading to WMI and motor impairment after ACMP. Furthermore, ferrostatin-1 reduced iron and ROS deposition to alleviate ferroptosis, thereafter reducing WMI to promote the recovery of motor function. The nuclear factor erythroid-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway was found to be involved in alleviating ferroptosis as seen with the administration of ferrostatin-1. The present study rationalizes that targeting ferroptosis to alleviate WMI is a feasible therapeutic strategy for managing ACMP.
Collapse
Affiliation(s)
- Shuhong Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Binyuan Xiong
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Yin Tian
- Department of Cardiology, The First People's Hospital of Zunyi, Zunyi, Guizhou, 133012, China
| | - Quan Hu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Ji Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Lin Chen
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Ruilie Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Mo Li
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Xin Zhou
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Tianxi Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Hongfei Ge
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| | - Anyong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563000, China.
| |
Collapse
|
32
|
Hu J, Cheng M, Jiang C, Liu L, He Z, Liu L, Yao Y, Li Z, Wang Q. Deferoxamine Mitigates Ferroptosis and Inflammation in Hippocampal Neurons After Subarachnoid Hemorrhage by Activating the Nrf2/TXNRD1 Axis. Mol Neurobiol 2024; 61:1044-1060. [PMID: 37676391 DOI: 10.1007/s12035-023-03525-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/17/2023] [Indexed: 09/08/2023]
Abstract
Ferroptosis is a distinct peroxidation-driven form of cell death tightly involved in subarachnoid hemorrhage (SAH). This study delved into the mechanism of deferoxamine (DFO, an iron chelator) in SAH-induced ferroptosis and inflammation. SAH mouse models were established by endovascular perforation method and injected intraperitoneally with DFO, or intraventricularly injected with the Nrf2 pathway inhibitor ML385 before SAH, followed by detection of neurological function, blood-brain barrier (BBB) permeability, and brain water content. Apoptotic level of hippocampal neurons, symbolic changes of ferroptosis, and levels of pro-inflammatory cytokines were assessed using TUNEL staining, Western blotting, colorimetry, and ELISA. The localization and expression of nuclear factor-erythroid 2-related factor 2 (Nrf2) were detected. HT22 cells were exposed to Hemin as in vitro SAH models and treated with FIN56 to induce ferroptosis, followed by evaluation of the effects of DFO on FIN56-treated HT22 cells. The regulation of Nrf2 in thioredoxin reductase 1 (TXNRD1) was analyzed by co-immunoprecipitation and Western blotting. Moreover, HT22 cells were treated with DFO and ML385 to identify the role of DFO in the Nrf2/TXNRD1 axis. DFO extenuated brain injury, and ferroptosis and inflammation in hippocampal neurons of SAH mice. Nrf2 localized at the CA1 region of hippocampal neurons, and DFO stimulated nuclear translocation of Nrf2 protein in hippocampal neurons of SAH mice. Additionally, DFO inhibited ferroptosis and inflammatory responses in FIN56-induced HT22 cells. Nrf2 positively regulated TXNRD1 protein expression. Indeed, DFO alleviated FIN56-induced ferroptosis and inflammation via activation of the Nrf2/TXNRD1 axis. DFO alleviated neurological deficits, BBB disruption, brain edema, and brain injury in mice after SAH by inhibiting hippocampal neuron ferroptosis via the Nrf2/TXNRD1 axis. DFO ameliorates SAH-induced ferroptosis and inflammatory responses in hippocampal neurons by activating the Nrf2/TXNRD1 axis.
Collapse
Affiliation(s)
- Junting Hu
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Meixiong Cheng
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Chonggui Jiang
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Ling Liu
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Zongze He
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Lingtong Liu
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Yuanpeng Yao
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China
| | - Zhili Li
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China.
| | - Qi Wang
- Department of Neurosurgery, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, Section 2, West 1St Ring Road, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
33
|
Fang D, Guo S, Wei B, Liu W, Li G, Li X, Liu J, Jin L, Duan C. Nrf-2 modulates excitability of hippocampal neurons by regulating ferroptosis and neuroinflammation after subarachnoid hemorrhage in rats. Brain Res Bull 2024; 207:110877. [PMID: 38215951 DOI: 10.1016/j.brainresbull.2024.110877] [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: 11/08/2023] [Revised: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 01/14/2024]
Abstract
Excitability of hippocampal neurons in subarachnoid hemorrhage (SAH) rats has not been well studied. The rat SAH model was applied in this study to explore the role of nuclear factor E2-related factor (Nrf-2) in the early brain injury of SAH. The neural excitability of CA1 pyramidal cells (PCs) in SAH rats was evaluated by using electrophysiology experiments. Ferroptosis and neuroinflammation were measured by ELISA, transmission electron microscopy and western blotting. Our results indicated that SAH induced neurological deficits, brain edema, ferroptosis, neuroinflammation and neural excitability in rats. Ferrostatin-1 treatment significantly decreased the expression and distribution of IL-1β, IL-6, IL-10, TGF-β and TNF-α. Inhibiting ferroptosis by ferrostatin-1 can attenuate neural excitability, neurological deficits, brain edema and neuroinflammation in SAH rats. Inhibiting the expression of Nrf-2 significantly increased the neural excitability and the levels of IL-1β, IL-6, IL-10, TGF-β and TNF-α in Fer-1-treated SAH rats. Taken together, inhibiting the Nrf-2 induces early brain injury, brain edema and the inflammatory response with increasing of neural excitability in Fer-1-treated SAH rats. These results have indicated that inhibiting ferroptosis, neuroinflammation and neural excitability attenuates early brain injury after SAH by regulating the Nrf-2.
Collapse
Affiliation(s)
- Dazhao Fang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China; Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Shenquan Guo
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Boyang Wei
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenchao Liu
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guangxu Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xifeng Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiahui Liu
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lei Jin
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuanzhi Duan
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
34
|
Dabbagh Ohadi MA, Maroufi SF, Mohammadi MR, Hosseini Siyanaki MR, Khorasanizadeh M, Kellner CP. Ferroptosis as a Therapeutic Target in Subarachnoid Hemorrhage. World Neurosurg 2024; 182:52-57. [PMID: 37979679 DOI: 10.1016/j.wneu.2023.11.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
Subarachnoid hemorrhage (SAH) is a cerebrovascular disorder with significant mortality and morbidity. Neural injury in SAH is mediated through a variety of pathophysiological processes. Currently available treatments are either nonspecific in targeting the basic pathophysiological mechanisms that result in neural damage in SAH, or merely focus on vasospasm. Ferroptosis is a type of programmed iron dependent cell death, which has received attention due to its possible role in neural injury in SAH. Herein, we review how intracellular iron overload mediates the production of reactive free radicals and lipid peroxidation through a variety of biochemical pathways in SAH. This in turn results in induction of ferroptosis, as well as exacerbation of vasospasm. We also discuss several therapeutic agents that have been shown to inhibit ferroptosis through targeting different steps of the process. Such agents have proven effective in ameliorating vasospasm, neural damage, and neurobehavioral outcomes in animal models of SAH. Human studies to test the safety and efficacy of intrathecal or parenteral administration of the inhibitors of ferroptosis in improving outcomes of SAH patients are warranted. There are currently a few ongoing clinical trials pursuing this therapeutic concept, the results of which will be critical to determine the value of ferroptosis as a novel therapeutic target in SAH.
Collapse
Affiliation(s)
- Mohammad Amin Dabbagh Ohadi
- Departments of Pediatric Neurosurgery Children's Medical Center Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Farzad Maroufi
- Neurosurgical Research Network (NRN), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Neurosurgery, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - MirHojjat Khorasanizadeh
- Department of Neurosurgery, Mount Sinai Hospital, Icahn School of Medicine, New York City, New York, USA.
| | - Christopher P Kellner
- Department of Neurosurgery, Mount Sinai Hospital, Icahn School of Medicine, New York City, New York, USA
| |
Collapse
|
35
|
Zhang J, Zhu Q, Peng Z, Li XJ, Ding PF, Gao S, Sheng B, Liu Y, Lu Y, Zhuang Z, Hang CH, Li W. Menaquinone-4 attenuates ferroptosis by upregulating DHODH through activation of SIRT1 after subarachnoid hemorrhage. Free Radic Biol Med 2024; 210:416-429. [PMID: 38042225 DOI: 10.1016/j.freeradbiomed.2023.11.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/07/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND Menaquinone-4(MK-4), the isoform of vitamin K2 in the brain, exerts neuroprotective effects against a variety of central nervous system disorders. This study aimed to demonstrate the anti-ferroptosis effects of MK-4 in neurons after SAH. METHODS A subarachnoid hemorrhage (SAH) model was prepared by endovascular perforation in mice. In vitro hemoglobin stimulation of primary cortical neurons mimicked SAH. MK-4, Brequinar (BQR, DHODH inhibitor), and Selisistat (SEL, SIRT1 inhibitor) were administered, respectively. Subsequently, WB, immunofluorescence was used to determine protein expression and localization, and transmission electron microscopy was used to observe neuronal mitochondrial structure while other indicators of ferroptosis were measured. RESULTS MK-4 treatment significantly upregulated the protein levels of DHODH; decreased GSH, PTGS2, NOX1, ROS, and restored mitochondrial membrane potential. Meanwhile, MK-4 upregulated the expression of SIRT1 and promoted its entry into the nucleus. BQR or SEL partially abolished the protective effect of MK-4 on, neurologic function, and ferroptosis. CONCLUSIONS Taken together, our results suggest that MK-4 attenuates ferroptosis after SAH by upregulating DHODH through the activation of SIRT1.
Collapse
Affiliation(s)
- Jiatong Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Qi Zhu
- Neurosurgical Institute, Nanjing University, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Zheng Peng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Xiao-Jian Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Peng-Fei Ding
- Neurosurgical Institute, Nanjing University, China; Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Sen Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Bin Sheng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Yang Liu
- Neurosurgical Institute, Nanjing University, China; Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
| | - Yue Lu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Zong Zhuang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China; Neurosurgical Institute, Nanjing University, China.
| |
Collapse
|
36
|
Hu Y, Gu X, Zhang Y, Ma W, Sun L, Wang C, Ren B. Adrenomedullin, transcriptionally regulated by vitamin D receptors, alleviates atherosclerosis in mice through suppressing AMPK-mediated endothelial ferroptosis. ENVIRONMENTAL TOXICOLOGY 2024; 39:199-211. [PMID: 37688783 DOI: 10.1002/tox.23958] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/31/2023] [Accepted: 08/20/2023] [Indexed: 09/11/2023]
Abstract
PURPOSE Vitamin D receptors (VDR) play important roles in cardiovascular, immune, metabolic and other functions. Activation of VDR may help improve endothelial dysfunction, atherosclerosis, vascular calcification, and cardiac hypertrophy. However, the specific target genes and mechanisms of VDR in improving Human Umbilical Vein Endothelial Cell (HUVEC) functions remain unclear. This study aims to investigate the function and mechanism of VDR in HUVECs. METHODS Endothelial dysfunction cell model was constructed by oxidized low-density lipoprotein (ox-LDL). An animal model of atherosclerosis was established in male homozygous Apoe-/- mice (6 weeks) on a high fat diet for 6 weeks. The relationship between VDR and adrenomedullin (ADM) was studied by bioinformatics analysis, ChIP, and luciferase reporter gene analysis. Endothelial cell function was evaluated by Transwell migration and Tube Formation tests. Ferroptosis was detected by measuring intracellular iron content, levels of oxidative stress markers, and ferroptosis related proteins. RESULTS Overexpression of VDR in HUVECs inhibits ox-LDL-induced endothelial dysfunction and ferroptosis. VDR binds to the ADM promoter sequence and regulates the transcription of ADM. Inhibition of ADM promotes ox-LDL-induced endothelial dysfunction and ferroptosis. ADM regulates ox-LDL-induced endothelial dysfunction and ferroptosis through the AMPK signaling pathway. Overexpression of VDR in Apoe-/- mice inhibited lipid deposition and plaque area in atherosclerotic mice. CONCLUSION VDR inhibits ox-LDL-induced endothelial dysfunction and ferroptosis by regulating ADM transcription and acting on AMPK signaling pathway. Overexpression of VDR in Apoe-/- mice reduced lipid deposition and plaque area in the thoracic aorta of atherosclerotic mice.
Collapse
Affiliation(s)
- Yanchao Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Xu Gu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Yan Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Weidong Ma
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Lijun Sun
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Congxia Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| | - Bincheng Ren
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Xi'an Jiaotong University, China
| |
Collapse
|
37
|
Abstract
Iron accumulation in the CNS occurs in many neurological disorders. It can contribute to neuropathology as iron is a redox-active metal that can generate free radicals. The reasons for the iron buildup in these conditions are varied and depend on which aspects of iron influx, efflux, or sequestration that help maintain iron homeostasis are dysregulated. Iron was shown recently to induce cell death and damage via lipid peroxidation under conditions in which there is deficient glutathione-dependent antioxidant defense. This form of cell death is called ferroptosis. Iron chelation has had limited success in the treatment of neurological disease. There is therefore much interest in ferroptosis as it potentially offers new drugs that could be more effective in reducing iron-mediated lipid peroxidation within the lipid-rich environment of the CNS. In this review, we focus on the molecular mechanisms that induce ferroptosis. We also address how iron enters and leaves the CNS, as well as the evidence for ferroptosis in several neurological disorders. Finally, we highlight biomarkers of ferroptosis and potential therapeutic strategies.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Fari Ryan
- Centre for Research in Neuroscience, and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Priya Jhelum
- Centre for Research in Neuroscience, and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Antje Kroner
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
38
|
Shao J, Meng Y, Yuan K, Wu Q, Zhu S, Li Y, Wu P, Zheng J, Shi H. RU.521 mitigates subarachnoid hemorrhage-induced brain injury via regulating microglial polarization and neuroinflammation mediated by the cGAS/STING/NF-κB pathway. Cell Commun Signal 2023; 21:264. [PMID: 37770901 PMCID: PMC10537158 DOI: 10.1186/s12964-023-01274-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/13/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND The poor prognosis of subarachnoid hemorrhage (SAH) is often attributed to neuroinflammation. The cGAS-STING axis, a cytoplasmic pathway responsible for detecting dsDNA, plays a significant role in mediating neuroinflammation in neurological diseases. However, the effects of inhibiting cGAS with the selective small molecule inhibitor RU.521 on brain injury and the underlying mechanisms after SAH are still unclear. METHODS The expression and microglial localization of cGAS following SAH were investigated with western blot analysis and immunofluorescent double-staining, respectively. RU.521 was administered after SAH. 2'3'-cGAMP, a second messenger converted by activated cGAS, was used to activate cGAS-STING. The assessments were carried out by adopting various techniques including neurological function scores, brain water content, blood-brain barrier permeability, western blot analysis, TUNEL staining, Nissl staining, immunofluorescence, morphological analysis, Morris water maze test, Golgi staining, CCK8, flow cytometry in the in vivo and in vitro settings. RESULTS Following SAH, there was an observed increase in the expression levels of cGAS in rat brain tissue, with peak levels observed at 24 h post-SAH. RU.521 resulted in a reduction of brain water content and blood-brain barrier permeability, leading to an improvement in neurological deficits after SAH. RU.521 had beneficial effects on neuronal apoptosis and microglia activation, as well as improvements in microglial morphology. Additionally, RU.521 prompted a shift in microglial phenotype from M1 to M2. We also noted a decrease in the production of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, and an increase in the level of the anti-inflammatory cytokine IL-10. Finally, RU.521 treatment was associated with improvements in cognitive function and an increase in the number of dendritic spines in the hippocampus. The therapeutic effects were mediated by the cGAS/STING/NF-κB pathway and were found to be abolished by 2'3'-cGAMP. In vitro, RU.521 significantly reduced apoptosis and neuroinflammation. CONCLUSION The study showed that SAH leads to neuroinflammation caused by microglial activation, which contributes to early brain injury. RU.521 improved neurological outcomes and reduced neuroinflammation by regulating microglial polarization through the cGAS/STING/NF-κB pathway in early brain injury after SAH. RU.521 may be a promising candidate for the treatment of neuroinflammatory pathology after SAH. Video Abstract.
Collapse
Affiliation(s)
- Jiang Shao
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yuxiao Meng
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Kaikun Yuan
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Qiaowei Wu
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Shiyi Zhu
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Yuchen Li
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Pei Wu
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China
| | - Jiaolin Zheng
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Xuefu Road 246#, Nangang District, Harbin, 150001, Heilongjiang Province, China.
| | - Huaizhang Shi
- Department of Neurosurgery, the First Affiliated Hospital of Harbin Medical University, Youzheng Street 23#, Nangang District, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
39
|
Wang Y, Wu S, Li Q, Sun H, Wang H. Pharmacological Inhibition of Ferroptosis as a Therapeutic Target for Neurodegenerative Diseases and Strokes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300325. [PMID: 37341302 PMCID: PMC10460905 DOI: 10.1002/advs.202300325] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/23/2023] [Indexed: 06/22/2023]
Abstract
Emerging evidence suggests that ferroptosis, a unique regulated cell death modality that is morphologically and mechanistically different from other forms of cell death, plays a vital role in the pathophysiological process of neurodegenerative diseases, and strokes. Accumulating evidence supports ferroptosis as a critical factor of neurodegenerative diseases and strokes, and pharmacological inhibition of ferroptosis as a therapeutic target for these diseases. In this review article, the core mechanisms of ferroptosis are overviewed and the roles of ferroptosis in neurodegenerative diseases and strokes are described. Finally, the emerging findings in treating neurodegenerative diseases and strokes through pharmacological inhibition of ferroptosis are described. This review demonstrates that pharmacological inhibition of ferroptosis by bioactive small-molecule compounds (ferroptosis inhibitors) could be effective for treatments of these diseases, and highlights a potential promising therapeutic avenue that could be used to prevent neurodegenerative diseases and strokes. This review article will shed light on developing novel therapeutic regimens by pharmacological inhibition of ferroptosis to slow down the progression of these diseases in the future.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care MedicineAerospace Center HospitalPeking University Aerospace School of Clinical MedicineBeijing100049P. R. China
| | - Shuang Wu
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhan430000P. R. China
| | - Qiang Li
- Department of NeurologyThe Affiliated Hospital of Chifeng UniversityChifeng024005P. R. China
| | - Huiyan Sun
- Chifeng University Health Science CenterChifeng024000P. R. China
| | - Hongquan Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060P. R. China
| |
Collapse
|
40
|
Yakubov E, Schmid S, Hammer A, Chen D, Dahlmanns JK, Mitrovic I, Zurabashvili L, Savaskan N, Steiner HH, Dahlmanns M. Ferroptosis and PPAR-gamma in the limelight of brain tumors and edema. Front Oncol 2023; 13:1176038. [PMID: 37554158 PMCID: PMC10406130 DOI: 10.3389/fonc.2023.1176038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/04/2023] [Indexed: 08/10/2023] Open
Abstract
Human malignant brain tumors such as gliomas are devastating due to the induction of cerebral edema and neurodegeneration. A major contributor to glioma-induced neurodegeneration has been identified as glutamate. Glutamate promotes cell growth and proliferation in variety of tumor types. Intriguently, glutamate is also an excitatory neurotransmitter and evokes neuronal cell death at high concentrations. Even though glutamate signaling at the receptor and its downstream effectors has been extensively investigated at the molecular level, there has been little insight into how glutamate enters the tumor microenvironment and impacts on metabolic equilibration until recently. Surprisingly, the 12 transmembrane spanning tranporter xCT (SLC7A11) appeared to be a major player in this process, mediating glutamate secretion and ferroptosis. Also, PPARγ is associated with ferroptosis in neurodegeneration, thereby destroying neurons and causing brain swelling. Although these data are intriguing, tumor-associated edema has so far been quoted as of vasogenic origin. Hence, glutamate and PPARγ biology in the process of glioma-induced brain swelling is conceptually challenging. By inhibiting xCT transporter or AMPA receptors in vivo, brain swelling and peritumoral alterations can be mitigated. This review sheds light on the role of glutamate in brain tumors presenting the conceptual challenge that xCT disruption causes ferroptosis activation in malignant brain tumors. Thus, interfering with glutamate takes center stage in forming the basis of a metabolic equilibration approach.
Collapse
Affiliation(s)
- Eduard Yakubov
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg, Germany
| | - Sebastian Schmid
- Department of Trauma, Orthopaedics, Plastic and Hand Surgery, University Hospital Augsburg, Augsburg, Germany
| | - Alexander Hammer
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg, Germany
- Center for Spine and Scoliosis Therapy, Malteser Waldkrankenhaus St. Marien, Erlangen, Germany
| | - Daishi Chen
- Department of Otorhinolaryngology, Shenzhen People's Hospital, Jinan University, Shenzhen, China
| | - Jana Katharina Dahlmanns
- Institute for Physiology and Pathophysiology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Ivana Mitrovic
- Department of Cardiac Surgery, Bogenhausen Hospital, Munich, Germany
| | | | - Nicolai Savaskan
- Department of Neurosurgery, University Medical School Hospital Universitätsklinikum Erlangen (UKER), Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Department of Public Health Neukölln, District Office Neukölln of Berlin Neukölln, Berlin, Germany
| | | | - Marc Dahlmanns
- Institute for Physiology and Pathophysiology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
41
|
Zhao Y, Luo Y, Liu Z, Chen Y, Wei L, Luo X, Zhou G, Lai J, Ji J, Lin Y, Liu J. Ferrostatin-1 ameliorates Bupivacaine-Induced spinal neurotoxicity in rats by inhibiting ferroptosis. Neurosci Lett 2023; 809:137308. [PMID: 37244447 DOI: 10.1016/j.neulet.2023.137308] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
Bupivacaine (BUP) has previously been shown to trigger neurotoxicity after spinal anesthesia. Further, ferroptosis has been implicated in the pathological processes associated with various central nervous system diseases. Although the impact of ferroptosis on BUP-induced neurotoxicity in the spinal cord has not been fully understood, this research aims to investigate this relationship in rats. Additionally, this study aims to determine whether ferrostatin-1 (Fer-1), a potent inhibitor of ferroptosis, can provide protection against BUP-induced spinal neurotoxicity. The experimental model for BUP-induced spinal neurotoxicity involved the administration of 5% bupivacaine through intrathecal injection. Then, the rats were randomized into the Control, BUP, BUP + Fer-1, and Fer-1 groups. BBB scores, %MPE of TFL, and H&E and Nissl stainings showed that intrathecal Fer-1 administration improved functional recovery, histological outcomes, and neural survival in BUP-treated rats. Moreover, Fer-1 has been found to alleviate the BUP-induced alterations related to ferroptosis, such as mitochondrial shrinkage and disruption of cristae, while also reducing the levels of malondialdehyde (MDA), iron, and 4-hydroxynonenal (4HNE). Fer-1 also inhibits the accumulation of reactive oxygen species (ROS) and restores the normal levels of glutathione peroxidase 4 (GPX4), cystine/glutamate transporter (xCT), and glutathione (GSH). Furthermore, double-immunofluorescence staining revealed that GPX4 is primarily localized in the neurons instead of microglia or astroglia in the spinal cord. In summary, we demonstrated that ferroptosis play a pivotal role in mediating BUP-induced spinal neurotoxicity, and Fer-1 ameliorated BUP-induced spinal neurotoxicity by reversing the underlying ferroptosis-related changes in rats.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China; Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, No.1 Maoyuan South Road, Nanchong 637000, Sichuan, China
| | - Yunpeng Luo
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China; Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou, China
| | - Ziru Liu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Yuanyuan Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Liling Wei
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Xi Luo
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Gang Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Jian Lai
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Jiemei Ji
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Yunan Lin
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Jingchen Liu
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China.
| |
Collapse
|
42
|
Deng X, Wu Y, Hu Z, Wang S, Zhou S, Zhou C, Gao X, Huang Y. The mechanism of ferroptosis in early brain injury after subarachnoid hemorrhage. Front Immunol 2023; 14:1191826. [PMID: 37266433 PMCID: PMC10229825 DOI: 10.3389/fimmu.2023.1191826] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/04/2023] [Indexed: 06/03/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is a cerebrovascular accident with an acute onset, severe disease characteristics, and poor prognosis. Within 72 hours after the occurrence of SAH, a sequence of pathological changes occur in the body including blood-brain barrier breakdown, cerebral edema, and reduced cerebrovascular flow that are defined as early brain injury (EBI), and it has been demonstrated that EBI exhibits an obvious correlation with poor prognosis. Ferroptosis is a novel programmed cell death mode. Ferroptosis is induced by the iron-dependent accumulation of lipid peroxides and reactive oxygen species (ROS). Ferroptosis involves abnormal iron metabolism, glutathione depletion, and lipid peroxidation. Recent study revealed that ferroptosis is involved in EBI and is significantly correlated with poor prognosis. With the gradual realization of the importance of ferroptosis, an increasing number of studies have been conducted to examine this process. This review summarizes the latest work in this field and tracks current research progress. We focused on iron metabolism, lipid metabolism, reduction systems centered on the GSH/GPX4 system, other newly discovered GSH/GPX4-independent antioxidant systems, and their related targets in the context of early brain injury. Additionally, we examined certain ferroptosis regulatory mechanisms that have been studied in other fields but not in SAH. A link between death and oxidative stress has been described. Additionally, we highlight the future research direction of ferroptosis in EBI of SAH, and this provides new ideas for follow-up research.
Collapse
Affiliation(s)
- Xinpeng Deng
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University, Ningbo, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yiwen Wu
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University, Ningbo, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Ziliang Hu
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Cixi, Zhejiang, China
| | - Shiyi Wang
- Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Shengjun Zhou
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University, Ningbo, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Chenhui Zhou
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University, Ningbo, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xiang Gao
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University, Ningbo, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Yi Huang
- Department of Neurosurgery, Ningbo First Hospital, Ningbo Hospital, Zhejiang University, Ningbo, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang, China
| |
Collapse
|
43
|
Zhang Z, Liu C, Zhou X, Zhang X. The Critical Role of Sirt1 in Subarachnoid Hemorrhages: Mechanism and Therapeutic Considerations. Brain Sci 2023; 13:brainsci13040674. [PMID: 37190639 DOI: 10.3390/brainsci13040674] [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: 02/22/2023] [Revised: 03/28/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
The subarachnoid hemorrhage (SAH) is an important cause of death and long-term disability worldwide. As a nicotinamide adenine dinucleotide-dependent deacetylase, silent information regulator 1 (Sirt1) is a multipotent molecule involved in many pathophysiological processes. A growing number of studies have demonstrated that Sirt1 activation may exert positive effects on SAHs by regulating inflammation, oxidative stress, apoptosis, autophagy, and ferroptosis. Thus, Sirt1 agonists may serve as potential therapeutic drugs for SAHs. In this review, we summarized the current state of our knowledge on the relationship between Sirt1 and SAHs and provided an updated overview of the downstream molecules of Sirt1 in SAHs.
Collapse
Affiliation(s)
- Zhonghua Zhang
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Cong Liu
- Department of Ophthalmology, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Xiaoming Zhou
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Xin Zhang
- Department of Neurosurgery, Jinling Hospital, Jinling School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
44
|
Jiao D, Xu J, Lou C, Luo Y, Ni C, Shen G, Fang M, Gong X. Quercetin alleviates subarachnoid hemorrhage-induced early brain injury via inhibiting ferroptosis in the rat model. Anat Rec (Hoboken) 2023; 306:638-650. [PMID: 36437694 DOI: 10.1002/ar.25130] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Abstract
Early brain injury (EBI) refers to a series of pathophysiological brain lesions that occur within 72 hr after subarachnoid hemorrhage (SAH), which is an extremely crucial factor in the poor prognosis of patients. In EBI, ferroptosis has been proven to cause neuronal death. Quercetin (QCT) is effective in deactivating reactive oxygen species (ROS), inhibiting lipid peroxidation, and even chelating iron, but its role in SAH remains unclear. In this study, the mortality rate, severity grade of SAH, brain water content (BWC), blood-brain barrier permeability, and neurological function of the rats were detected. Moreover, mitochondrial morphology in cortical neurons were observed and their sizes were subsequently quantified. The levels of lipid peroxidation on glutathione and malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) were determined, whereas the protein expressions of glutathione peroxidase 4 (GPX4), SLC7A11 (xCT), transferrin receptor 1 (TfR1), and ferroportin-1 (FPN1) were analyzed by western immunoblotting. The neurodegeneration involved in EBI was investigated by fluoro-Jade C staining, while iron staining was utilized to measure iron content. Our results showed that inhibition of ferroptosis by QCT could suppress EBI and improve neurological function in SAH rats. QCT increased the expression levels of GPX4, xCT, and FPN1, while downregulated TfR1, and exerted protective effects on neurons as well as alleviated iron accumulation and lipid peroxidation in the cortex of SAH rats. In conclusion, our study revealed that QCT might alleviate the EBI by inhibiting ferroptosis in SAH rats.
Collapse
Affiliation(s)
- Dian Jiao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Jianmiao Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Chengjian Lou
- Department of Neurosurgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Yuhuan Luo
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengtao Ni
- Graduate School, Bengbu Medical College, Bengbu, China
| | - Guanghong Shen
- The Affiliated People's Hospital of Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China
| | - Marong Fang
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangyang Gong
- Rehabilitation Medicine Center, Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
45
|
Pharmacological Modulations of Nrf2 and Therapeutic Implications in Aneurysmal Subarachnoid Hemorrhage. Molecules 2023; 28:molecules28041747. [PMID: 36838735 PMCID: PMC9963186 DOI: 10.3390/molecules28041747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
An aneurysmal subarachnoid hemorrhage (aSAH) is a subtype of stroke with high morbidity and mortality. The main causes of a poor prognosis include early brain injury (EBI) and delayed vasospasm, both of which play a significant role in the pathophysiological process. As an important mechanism of EBI and delayed vasospasm, oxidative stress plays an important role in the pathogenesis of aSAH by producing reactive oxygen species (ROS) through the mitochondria, hemoglobin, or enzymatic pathways in the early stages of aSAH. As a result, antioxidant therapy, which primarily targets the Nrf2-related pathway, can be employed as a potential strategy for treating aSAH. In the early stages of aSAH development, increasing the expression of antioxidant enzymes and detoxifying enzymes can relieve oxidative stress, reduce brain damage, and improve prognosis. Herein, the regulatory mechanisms of Nrf2 and related pharmacological compounds are reviewed, and Nrf2-targeted drugs are proposed as potential treatments for aSAH.
Collapse
|
46
|
Liu X, Du Y, Liu J, Cheng L, He W, Zhang W. Ferrostatin-1 alleviates cerebral ischemia/reperfusion injury through activation of the AKT/GSK3β signaling pathway. Brain Res Bull 2023; 193:146-157. [PMID: 36596364 DOI: 10.1016/j.brainresbull.2022.12.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 01/02/2023]
Abstract
Ischemic stroke is the major cause of disability and death worldwide, but post-stroke neuronal death and related mechanisms remain unclear. Ferroptosis, a newly identified type of regulated cell death, has been shown to be associated with neurological disorders, yet the exact relationship between ferroptosis and ischemic stroke has not been elucidated. The purpose of this study is to investigate the effects of ferroptosis-specific inhibitor ferrostatin-1 (Fer-1) on neuronal injury after cerebral ischemia/reperfusion (I/R) and the underlying mechanism. In this study, we demonstrated that ferroptosis does occur in the stroke model. We found that Fer-1 reduced the levels of iron and malondialdehyde, and increased the content of glutathione and the expression of solute carrier family 7 member 11 and glutathione peroxidase 4 in cerebral I/R models. Additionally, Fer-1 significantly reduced the infarct volume and improved neurobehavioral outcomes. Moreover, we found that Fer-1 increased the levels of phosphorylated AKT and GSK3β following cerebral I/R. To further investigate the functional role of the AKT in the neuroprotective effects of Fer-1, MCAO models and oxygen-glucose deprivation-induced HT22 cells were pretreated with the AKT inhibitor MK-2206 before treatment with Fer-1 and the protective effects of Fer-1 were reversed. In conclusion, Fer-1 has protective effects on cerebral I/R injury by activating the AKT/GSK3β pathway, indicating that ferroptosis may become a novel target in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Xinyao Liu
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Yue Du
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Jian Liu
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Linggang Cheng
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Wen He
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Wei Zhang
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
47
|
Pan F, Xu W, Ding J, Wang C. Elucidating the progress and impact of ferroptosis in hemorrhagic stroke. Front Cell Neurosci 2023; 16:1067570. [PMID: 36713782 PMCID: PMC9874704 DOI: 10.3389/fncel.2022.1067570] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
Hemorrhagic stroke is a devastating cerebrovascular disease with high morbidity and mortality, for which effective therapies are currently unavailable. Based on different bleeding sites, hemorrhagic stroke can be generally divided into intracerebral hemorrhage (ICH) and subarachnoid hemorrhage (SAH), whose pathogenesis share some similarity. Ferroptosis is a recently defined programmed cell deaths (PCDs), which is a critical supplement to the hypothesis on the mechanism of nervous system injury after hemorrhagic stroke. Ferroptosis is characterized by distinctive morphological changes of mitochondria and iron-dependent accumulation of lipid peroxides. Moreover, scientists have successfully demonstrated the involvement of ferroptosis in animal models of ICH and SAH, indicating that ferroptosis is a promising target for hemorrhagic stroke therapy. However, the studies on ferroptosis still faces a serious of technical and theoretical challenges. This review systematically elaborates the role of ferroptosis in the pathogenesis of hemorrhagic stroke and puts forward some opinions on the dilemma of ferroptosis research.
Collapse
Affiliation(s)
- Feixia Pan
- Department of Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Weize Xu
- Department of Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jieying Ding
- Department of Pharmacy, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chencen Wang
- Department of Pediatrics, The First People’s Hospital of Yongkang Affiliated to Hangzhou Medical College, Jinhua, China,*Correspondence: Chencen Wang,
| |
Collapse
|
48
|
Wang J, Yang S, Jing G, Wang Q, Zeng C, Song X, Li X. Inhibition of ferroptosis protects sepsis-associated encephalopathy. Cytokine 2023; 161:156078. [PMID: 36401983 DOI: 10.1016/j.cyto.2022.156078] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is a serious and common complication of sepsis. To study the ferroptosis in the pathogenesis of SAE and demonstrate the protection effect of ferroptosis resistance, cognitive function, neurological deficits, blood-brain barrier integrity and neuroinflammation were detected. SAE model was established by cecal ligation and puncture (CLP) in mice and an in vitro model was created by introducing LPS to HT22 cells. Ferroptosis inducer Fe-citrate (Fe) and ferroptosis inhibitor ferrostatin-1 (Fer-1) was post-treated in the models, respectively. SAE caused ferroptosis, as evidenced by an increase in reactive oxygen species (ROS), iron content and malondialdehyde (MDA) and a decrease in glutathione (GSH) level, as well as changes in the expression of ferroptosis-related proteins as acyl-CoA synthetase long-chain family member 4 (ACSL4), glutathione peroxidase 4 (GPX4), and cystine-glutamate antiporter (SLC7A11), and harmed mitochondrial function. In contrast, inhibiting ferroptosis with Fer-1 attenuated ferroptosis. Meanwhile, Fer-1 attenuated neurologic severity score, learning and memory impairment, Fluoro-Jade C (FJC) staining, and decreased Evans Blue (EB) extravasation, microglia activation and TNF-α and IL-1β production following SAE. The benefit of Fer-1 was diminished by ferroptosis inducer Fe. In addition, Fer-1 up-regulated the nuclear factor erythroid-2-related factor 2 (Nrf2)/ heme oxygenase-1(HO-1) signaling axis both in vivo and in vitro. In conclusion, our study revealed that Fer-1 might inhibit feroptosis in neurons by triggering the Nrf2/OH-1 pathway, thereby providing a therapeutic solution for SAE.
Collapse
Affiliation(s)
- Jin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shuhua Yang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guoqing Jing
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qingyuan Wang
- Department of Anesthesiology, the People's Hospital of Tuanfeng, Huanggang, Hubei, China
| | - Cheng Zeng
- Institute of Hepatobiliary Diseases of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei, China.
| | - Xuemin Song
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Xinyi Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
49
|
Xu Y, Li K, Zhao Y, Zhou L, Liu Y, Zhao J. Role of Ferroptosis in Stroke. Cell Mol Neurobiol 2023; 43:205-222. [PMID: 35102454 PMCID: PMC11415219 DOI: 10.1007/s10571-022-01196-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/18/2022] [Indexed: 01/07/2023]
Abstract
Stroke is a common and serious nervous system disease caused by the rupture or blockage of the cardiovascular system. It causes millions of deaths and disabilities every year, which is a huge burden on humanity. It may be induced by thrombosis, hypertension, hyperlipidemia, hyperglycemia, smoking, advanced age and so on. According to different causes, stroke can be generally divided into hemorrhagic stroke and ischemic stroke, whose pathogenesis and treatment are quite different. Ferroptosis is a new type of cell death first defined in 2012, which is characterized by non-apoptotic, iron-dependent, and over-accumulated lipid peroxides. Excess lipid reactive oxygen species produced during ferroptosis eventually leads to oxidative cell death. Ferroptosis has been shown to occur and play an important role in tumors, neurological diseases, kidney injury, and ischemia-reperfusion injury. Ferroptosis is also closely related to the pathogenesis of stroke. Moreover, scientists have successfully intervened in the process of stroke in animal models by regulating ferroptosis, indicating that ferroptosis is a new potential target for the treatment of stroke. This paper systematically summarizes the involvement and role of ferroptosis in the pathogenesis of stroke and predicts the potential of ferroptosis in the treatment of stroke. Ferroptosis in stroke. Stroke induces iron overload and lipid metabolism disorders. Elevated iron catalyzes lipid peroxidation and eventually triggers ferroptosis. Conversely, the GSH/GPX4 pathway, as well as CoQ10, Fer-1, and Lip-1, inhibits lipid peroxidation and, thus, alleviates ferroptosis. GSH glutathione; GPX4 glutathione peroxidase 4; CoQ10 coenzyme Q10; Lip-1 liproxstatin-1; Fer-1 ferostatin-1.
Collapse
Affiliation(s)
- Yunfei Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410008, Hunan, China
| | - Kexin Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410008, Hunan, China
| | - Yao Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410008, Hunan, China
| | - Lin Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410008, Hunan, China
| | - Ying Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, 410008, Hunan, China.
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, 410008, Hunan, China.
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410008, Hunan, China.
| | - Jie Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
50
|
Qiu X, Tao Q, Zhang L, Kuang C, Xie Y, Zhang L, Yin S, Peng J, Jiang Y. Deletion of Bak1 alleviates microglial necroptosis and neuroinflammation after experimental subarachnoid hemorrhage. J Neurochem 2022; 164:829-846. [PMID: 36583235 DOI: 10.1111/jnc.15751] [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: 06/09/2022] [Revised: 11/18/2022] [Accepted: 12/06/2022] [Indexed: 12/31/2022]
Abstract
Microglial necroptosis exacerbates neurodegenerative diseases, central nervous system (CNS) injury, and demonstrates a proinflammatory process, but its contribution to subarachnoid hemorrhage (SAH) is poorly characterized. BCL-2 homologous antagonist-killer protein (Bak1), a critical regulatory molecule of endogenous apoptosis, can be involved in the pathologic process of necroptosis by regulating mitochondrial permeability. In this study, we revealed microglia undergo necroptosis after SAH in vivo and vitro. Western blot revealed that Bak1 was elevated at 24 h after SAH. Knocked down of Bak1 by adeno-associated virus attenuates microglial necroptosis, alleviates neuroinflammation, and improves neurologic function after SAH in mice. Furthermore, oxyhemoglobin (10 μM) induced necroptosis in BV2 microglia, increasing Bak1 expression and mediating proinflammatory phenotype transformation, exacerbating oxidative stress and neuroinflammation. Abrogating BV2 Bak1 could reduce necroptosis by down-regulating the expression of phosphorylated pseudokinase mixed lineage kinase domain-like protein (p-MLKL), then down-regulating proinflammatory phenotype gene expression. RNA-Seq showed that disrupting BV2 Bak1 down-regulates multiple immune and inflammatory pathways and ameliorates cell injury by elevating thrombospondin 1 (THBS1) expression. In summary, we identified a critical regulatory role for Bak1 in microglial necroptosis and neuroinflammation after SAH. Bak1 is expected to be a potential target for the treatment strategy of SAH.
Collapse
Affiliation(s)
- Xiancheng Qiu
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Neurosurgery, Shifang City People's Hospital, Shifang, China
| | - Qianke Tao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lihan Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chenghao Kuang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China
| | - Yuke Xie
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China
| | - Lifang Zhang
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shigang Yin
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|