1
|
Fernández-Pérez I, Jiménez-Balado J, Macias-Gómez A, Suárez-Pérez A, Vallverdú-Prats M, Pérez-Giraldo A, Viles-García M, Peris-Subiza J, Vidal-Notari S, Giralt-Steinhauer E, Guisado-Alonso D, Esteller M, Rodriguez-Campello A, Jiménez-Conde J, Ois A, Cuadrado-Godia E. Blood DNA Methylation Analysis Reveals a Distinctive Epigenetic Signature of Vasospasm in Aneurysmal Subarachnoid Hemorrhage. Transl Stroke Res 2025; 16:715-727. [PMID: 38649590 DOI: 10.1007/s12975-024-01252-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: 02/27/2024] [Revised: 03/28/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
Vasospasm is a potentially preventable cause of poor prognosis in patients with aneurysmal subarachnoid hemorrhage (aSAH). Epigenetics might provide insight on its molecular mechanisms. We aimed to analyze the association between differential DNA methylation (DNAm) and development of vasospasm. We conducted an epigenome-wide association study in 282 patients with aSAH admitted to our hospital. DNAm was assessed with the EPIC Illumina chip (> 850 K CpG sites) in whole-blood samples collected at hospital admission. We identified differentially methylated positions (DMPs) at the CpG level using Cox regression models adjusted for potential confounders, and then we used the DMP results to find differentially methylated regions (DMRs) and enriched biological pathways. A total of 145 patients (51%) experienced vasospasm. In the DMP analysis, we identified 31 CpGs associated with vasospasm at p-value < 10-5. One of them (cg26189827) was significant at the genome-wide level (p-value < 10-8), being hypermethylated in patients with vasospasm and annotated to SUGCT gene, mainly expressed in arteries. Region analysis revealed 13 DMRs, some of them annotated to interesting genes such as POU5F1, HLA-DPA1, RUFY1, and CYP1A1. Functional enrichment analysis showed the involvement of biological processes related to immunity, inflammatory response, oxidative stress, endothelial nitric oxide, and apoptosis. Our findings show, for the first time, a distinctive epigenetic signature of vasospasm in aSAH, establishing novel links with essential biological pathways, including inflammation, immune responses, and oxidative stress. Although further validation is required, our results provide a foundation for future research into the complex pathophysiology of vasospasm.
Collapse
Affiliation(s)
- Isabel Fernández-Pérez
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | - Joan Jiménez-Balado
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain.
| | - Adrià Macias-Gómez
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | - Antoni Suárez-Pérez
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | - Marta Vallverdú-Prats
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | | | - Marc Viles-García
- Neuroradiology Department, Hospital del Mar, Barcelona, Catalunya, Spain
| | | | | | - Eva Giralt-Steinhauer
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| | - Daniel Guisado-Alonso
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Research Institute Against Leukemia Josep Carreras, Badalona, Catalunya, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalunya, Spain
| | - Ana Rodriguez-Campello
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| | - Jordi Jiménez-Conde
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| | - Angel Ois
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| | - Elisa Cuadrado-Godia
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| |
Collapse
|
2
|
Saadh MJ, Ahmed HH, Sanghvi G, Bin Awang Isa MZ, Singh P, Kaur K, Kumar MR, Husseen B. Recent advances in the delivery of microRNAs via exosomes derived from MSCs, and their role in regulation of ferroptosis. Pathol Res Pract 2025; 270:155984. [PMID: 40315562 DOI: 10.1016/j.prp.2025.155984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025]
Abstract
Mesenchymal stem cell (MSC) therapy, with its unique properties, has garnered interest in cancer treatment. Exosomes (EXOs)-derived from MSC retain the paracrine components of MSCs and demonstrate increased stability, minimal immunogenicity, and low risk of unintended tumorigenesis. Enhanced endocytosis methods make them versatile delivery vehicles for therapeutic cargo. MSC-EXOs can either promote or inhibit carcinogenesis, mediated by paracrine factors and various RNA molecules, particularly microRNAs (miRNAs). The prospect of using MSC-EXOs as a delivery tool for antitumor miRNAs in solid tumor therapy is promising. Exosomes' intrinsic tumor-targeting abilities and low immunogenicity make them ideal for delivering miRNAs, which have shown potential as cancer therapeutics. miRNAs within MSC-EXOs molecules can stimulate tumor growth or induce non-apoptotic cell death pathways, such as ferroptosis, depending on context. Ferroptosis is a kind of controlled cell death that is associated with the pathophysiology of several illnesses and includes iron metabolism. There is growing evidence that miRNAs carried by exosomes derived from MSCs may control ferroptosis in tumor cells by altering key genes related to antioxidant defense, lipid peroxidation, and iron metabolism. Understanding their complex mechanisms in the tumor microenvironment and optimizing their cargo are critical steps toward harnessing their full therapeutic potential. This review provides a comprehensive overview of MSC-EXOs and their role in cancer treatment. We also discuss the potential of MSC-EXOs as delivery vehicles for miRNAs to enhance therapeutic efficacy, as well as the role of exosomal miRNAs in the induction of ferroptosis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | | | - Priyanka Singh
- NIMS School of Allied Sciences and Technology, NIMS University, Jaipur, Rajasthan 303121, India
| | - Kiranjeet Kaur
- Chandigarh Pharmacy College, Chandigarh Group of colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Beneen Husseen
- Medical laboratory technique college, the Islamic University, Najaf, Iraq; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
3
|
Xiao W, Yike W, Gongwen L, Youjia X. Ferroptosis-mediated immune responses in osteoporosis. J Orthop Translat 2025; 52:116-125. [PMID: 40271049 PMCID: PMC12017889 DOI: 10.1016/j.jot.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Osteoporosis is a common systemic metabolic disease, characterized by decreased bone mass and susceptibility to fragility fractures, often associated with aging, menopause, genetics, and immunity. Ferroptosis plays an underestimated yet crucial role in the further impact of immune function changes on osteoporosis. Cell ferroptosis can induce alterations in immune function, subsequently influencing bone metabolism. In this context, this review summarizes several mechanisms of ferroptosis and introduces the latest insights on how ferroptosis regulates immune responses, exploring the interactions between ferroptosis and other mechanisms such as oxidative stress, inflammation, etc. This review elucidates potential treatment strategies for osteoporosis, emphasizing the promising potential of ferroptosis as an emerging target in the treatment of osteoporosis. In conclusion, preparations related to ferroptosis exhibit substantial clinical promise for enhancing bone mass restoration. The translational potential of this article: This review elucidates a nuanced conversation between the immune system and osteoporosis, with ferroptosis serving as the connecting link. These findings underscore the potential of ferroptosis inhibition as a therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Wang Xiao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wang Yike
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liu Gongwen
- Department of Orthopaedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xu Youjia
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Liu Q, Han Z, Li T, Meng J, Zhu C, Wang J, Wang J, Zhang Z, Wu H. Microglial HO-1 aggravates neuronal ferroptosis via regulating iron metabolism and inflammation in the early stage after intracerebral hemorrhage. Int Immunopharmacol 2025; 147:113942. [PMID: 39740507 DOI: 10.1016/j.intimp.2024.113942] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
Heme oxygenase 1 (HO-1), an enzyme involved in heme catabolism, has been shown upregulated in microglia cells and plays a critical roles in neurological damages after intracerebral hemorrhage (ICH). However, the mechanisms by which HO-1 mediates the neuronal damages are still obscure. Here, our findings demonstrate that HO-1 over-expression exacerbates the pro-inflammatory response of microglia and induces neuronal ferroptosis through promoting intracellular iron deposition in the ICH model both in vitro and in vivo. Furthermore, in the co-cultured ICH model in vitro, we verify that HO-1 over-expression disrupts the balance of iron metabolism in microglia, which increases the iron efflux to the extracellular space and promotes iron ion uptake in neurons, leading to lipid peroxidation injury and further contributing to neuronal ferroptosis. Moreover, the specific ferroptosis inhibitor Ferrostatin-1 (Fer-1) treatment could mitigate the damages in the co-cultured HT22 cells that caused by HO-1 over-expression in microglia, and improve the neurological function in the ICH model in mice. By shedding light on the mechanisms of aggravating neuronal ferroptosis due to HO-1 overexpression in the early stages after ICH, our study provides insights into the potential therapy of targeting HO-1 to treat ICH.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ziyi Han
- College of Medical Laboratory Technology, Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Tao Li
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jincheng Meng
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Chenwei Zhu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jian Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Zhen Zhang
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| | - He Wu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
5
|
Huang S, Li W, Wang D, Feng H, Wang B, Dong X, Zhao W, Liu D, Wang Y. Maternal exposure to deltamethrin during pregnancy and lactation impairs hippocampal learning and memory function of male offspring by ferroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117729. [PMID: 39818137 DOI: 10.1016/j.ecoenv.2025.117729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
Deltamethrin (DM), a broad-spectrum insecticide, is widely used in the world. It can exert direct action on the central nervous system to produce neurotoxicity. Exposure to DM can lead to iron metabolism disorder, oxidative stress and learning and memory dysfunction. In our study, pregnant Wistar rats were randomly divided into four groups and gavaged at doses of 0, 1, 4 or 10 mg/kg/d DM from gestational day (GD) 0 to postnatal day (PND) 21. We used behavioral experiments and Nissl staining to observe the hippocampal development and learning and memory function of male offspring. In order to further confirm the regulation mechanisms of DM, we detected ferrous ion, oxidative stress, ferroptosis related proteins, phospholipase C (PL-C)/inositol triphosphate 3 receptor (IP3R) signaling pathway, intracellular Ca2+ and calcineurin (CaN) content in vivo. In vitro,we selected HT-22 cells to be exposed to DM under the intervention of ferrostatin-1 and pifithrin-α. Our results showed that maternal exposure to DM reduced T-maze correctness and the number of hippocampal neurons, and increased shuttle box passive avoidance rate. Moreover, maternal exposure to DM increased the expression of ferrous ion, malondialdehyde (MDA) and prostaglandin-endoperoxide synthase 2 (PTGS2) protein, and decreased the glutathione (GSH) level in the hippocampus, which was contributed to ferroptosis by p53-mediated solute carrier family 7 member 11 (SLC7A11)/glutathione peroxidase 4 (GPX4) axis in the male offspring. Furthermore, the ferroptosis caused by DM exposure could active PL-C/IP3R signaling pathway and increase the intracellular Ca2+ and CaN level, leading to an imbalance of calcium homeostasis in the hippocampus. Thus, maternal exposure to DM during pregnancy and lactation could impair hippocampal learning and memory function of male offspring by p53-mediated ferroptosis.
Collapse
Affiliation(s)
- Shasha Huang
- Yongchuan Center for Disease Control and Prevention, Chongqing, China; Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Wanqi Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, China; Tai'an Center for Disease Control and Prevention, Shandong, China
| | - Dengke Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Huiwen Feng
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Bo Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Xinyu Dong
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Wei Zhao
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Dan Liu
- NHC Key Laboratory of Congenital Malformation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Yuan Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, Liaoning, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
6
|
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
|
7
|
Hao J, Wang T, Cao C, Li X, Li H, Gao H, Li J, Shen H, Chen G. LPCAT3 exacerbates early brain injury and ferroptosis after subarachnoid hemorrhage in rats. Brain Res 2024; 1832:148864. [PMID: 38484924 DOI: 10.1016/j.brainres.2024.148864] [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: 10/31/2023] [Revised: 02/19/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
AIMS Lysophosphatidylcholine acyltransferase 3 (LPCAT3) is known to play a pivotal role in lipid metabolism, but its role in the early brain injury (EBI) following subarachnoid hemorrhage (SAH) remains unclear. This study provides insights into LPCAT3 expression alterations and functional implications in EBI following SAH. METHODS SAH models of adult male Sprague-Dawley (SD) rats were established by intravascular perforation. Lentivirus vectors were administered by intracerebroventricular injection (i.c.v.) to either induce LPCAT3 overexpression or knockdown 14 days before SAH induction. Western blot, immunofluorescence, Nissl staining, MDA detection, ROS detection, iron content detection, and short-term and long-term neurobehavioral tests were performed to investigate the effects of regulated-LPCAT3 after SAH. RESULTS LPCAT3 levels were found to be significantly elevated in SAH. Suppression of LPCAT3 expression via shRNA improved oxidative stress, reduced brain edema, alleviated behavioral and cognitive deficits following SAH and decreased neuronal death, while upregulating LPCAT3 expression showed opposing effects. CONCLUSION LPCAT3 is involved in SAH-induced EBI and associated with ferroptosis. Our findings provide a referential basis for potential therapeutic interventions aimed at alleviating EBI following SAH.
Collapse
Affiliation(s)
- Jiahui Hao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Tong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Intensive Care Unit, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China; Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Heng Gao
- Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China
| | - Jinquan Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
8
|
Cui M, Chen F, Shao L, Wei C, Zhang W, Sun W, Wang J. Mesenchymal stem cells and ferroptosis: Clinical opportunities and challenges. Heliyon 2024; 10:e25251. [PMID: 38356500 PMCID: PMC10864896 DOI: 10.1016/j.heliyon.2024.e25251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Objective This review discusses recent experimental and clinical findings related to ferroptosis, with a focus on the role of MSCs. Therapeutic efficacy and current applications of MSC-based ferroptosis therapies are also discussed. Background Ferroptosis is a type of programmed cell death that differs from apoptosis, necrosis, and autophagy; it involves iron metabolism and is related to the pathogenesis of many diseases, such as Parkinson's disease, cancers, and liver diseases. In recent years, the use of mesenchymal stem cells (MSCs) and MSC-derived exosomes has become a trend in cell-free therapies. MSCs are a heterogeneous cell population isolated from a diverse range of human tissues that exhibit immunomodulatory functions, regulate cell growth, and repair damaged tissues. In addition, accumulating evidence indicates that MSC-derived exosomes play an important role, mainly by carrying a variety of bioactive substances that affect recipient cells. The potential mechanism by which MSC-derived exosomes mediate the effects of MSCs on ferroptosis has been previously demonstrated. This review provides the first overview of the current knowledge on ferroptosis, MSCs, and MSC-derived exosomes and highlights the potential application of MSCs exosomes in the treatment of ferroptotic conditions. It summarizes their mechanisms of action and techniques for enhancing MSC functionality. Results obtained from a large number of experimental studies revealed that both local and systemic administration of MSCs effectively suppressed ferroptosis in injured hepatocytes, neurons, cardiomyocytes, and nucleus pulposus cells and promoted the survival and regeneration of injured organs. Methods We reviewed the role of ferroptosis in related tissues and organs, focusing on its characteristics in different diseases. Additionally, the effects of MSCs and MSC-derived exosomes on ferroptosis-related pathways in various organs were reviewed, and the mechanism of action was elucidated. MSCs were shown to improve the disease course by regulating ferroptosis.
Collapse
Affiliation(s)
- Mengling Cui
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Fukun Chen
- Department of Radiology, Kunming Medical University & the Third Affiliated Hospital, Kunming, Yunnan, 650101, PR China
| | - Lishi Shao
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Chanyan Wei
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Weihu Zhang
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Wenmei Sun
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Jiaping Wang
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
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: 2] [Impact Index Per Article: 2.0] [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
|
12
|
Cao C, Lu T, Cheng Q, Cui G, Wang Z, Li X, Li H, Gao H, Shen H, Sun Q. Restoring System xc- activity by xCT overexpression inhibited neuronal ferroptosis and improved neurological deficits after experimental subarachnoid hemorrhage. Brain Res 2023; 1820:148556. [PMID: 37648093 DOI: 10.1016/j.brainres.2023.148556] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Ferroptosis is an important therapeutic target to alleviate early brain injury (EBI) after subarachnoid hemorrhage (SAH), yet the mechanism of neuronal ferroptosis after SAH remains unclear. System xc- dysfunction is one of the key pathways to induce ferroptosis. System xc- activity is mainly regulated by the expression of xCT. This study was designed to investigate the effect of xCT expression and System xc- activity on ferroptosis and EBI in an experimental SAH model both in vitro and in vivo. METHODS SAH was induced in adult male Sprague-Dawley rats by injecting autologous blood into the prechiasmatic cistern. Primary neurons treated with oxyhemoglobin (10 µM) were used to mimic SAH in vitro. Plasmid transfection was used to induce xCT overexpression. Western blotting, immunofluorescence staining, measurement of cystine uptake, enzyme-linked immunosorbent assay, transmission electron microscopy, Nissl staining, and a series of neurobehavioral tests were conducted to explore the role of xCT and System xc- activity in ferroptosis and EBI after SAH. RESULTS We found that System xc- dysfunction induced ferroptosis and exacerbated EBI after SAH in rats. xCT deficiency after SAH resulted in System xc- dysfunction, weakened neuronal antioxidant capacity and activated neuronal ferroptosis. xCT overexpression improved neuronal antioxidant capacity and inhibited neuronal ferroptosis by restoring System xc- activity. Rats with xCT overexpression after SAH presented with attenuated brain edema and inflammation, increased neuronal survival, and ameliorated neurological deficits. CONCLUSIONS Our study revealed that restoring System xc- activity by xCT overexpression inhibited neuronal ferroptosis and EBI and improved neurological deficits after SAH.
Collapse
Affiliation(s)
- Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Intensive Care Unit, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China; Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China.
| | - Ting Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Qian Cheng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Gang Cui
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Heng Gao
- Department of Brain Center, The Affiliated Jiangyin Hospital of Nantong University, Jiangyin City 214400, Jiangsu Province, China.
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| |
Collapse
|
13
|
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
|
14
|
Xu R, Wang W, Zhang W. Ferroptosis and the bidirectional regulatory factor p53. Cell Death Discov 2023; 9:197. [PMID: 37386007 DOI: 10.1038/s41420-023-01517-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023] Open
Abstract
Ferroptosis is a type of regulated cell death characterized by iron-mediated lipid peroxidation, in contrast with apoptosis, autophagy, and necrosis. It can be triggered by many pathological processes, including cellular metabolism, tumors, neurodegenerative diseases, cardiovascular diseases, and ischemia-reperfusion injuries. In recent years, ferroptosis has been discovered to be associated with p53. P53 is a tumor suppressor protein with multiple and powerful functions in cell cycle arrest, senescence, cell death, repair of DNA damage, and mitophagy. Emerging evidence shows that ferroptosis plays a crucial role in tumor suppression by p53. P53 functions as a key bidirectional regulator of ferroptosis by adjusting metabolism of iron, lipids, glutathione peroxidase 4, reactive oxygen species, and amino acids via a canonical pathway. In addition, a noncanonical pathway of p53 that regulates ferroptosis has been discovered in recent years. The specific details require to be further clarified. These mechanisms provide new ideas for clinical applications, and translational studies of ferroptosis have been performed to treat various diseases.
Collapse
Affiliation(s)
- Ren Xu
- Pulmonary and Critical Care Medicine Department, First Hospital of Jiliwn University, 130021, Changchun, China
| | - Wanning Wang
- Nephrology Department, First Hospital of Jilin University, 130021, Changchun, China
| | - Wenlong Zhang
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, 130033, Changchun, China.
| |
Collapse
|
15
|
Jia B, Li J, Song Y, Luo C. ACSL4-Mediated Ferroptosis and Its Potential Role in Central Nervous System Diseases and Injuries. Int J Mol Sci 2023; 24:10021. [PMID: 37373168 DOI: 10.3390/ijms241210021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
As an iron-dependent regulated form of cell death, ferroptosis is characterized by iron-dependent lipid peroxidation and has been implicated in the occurrence and development of various diseases, including nervous system diseases and injuries. Ferroptosis has become a potential target for intervention in these diseases or injuries in relevant preclinical models. As a member of the Acyl-CoA synthetase long-chain family (ACSLs) that can convert saturated and unsaturated fatty acids, Acyl-CoA synthetase long-chain familymember4 (ACSL4) is involved in the regulation of arachidonic acid and eicosapentaenoic acid, thus leading to ferroptosis. The underlying molecular mechanisms of ACSL4-mediated ferroptosis will promote additional treatment strategies for these diseases or injury conditions. Our review article provides a current view of ACSL4-mediated ferroptosis, mainly including the structure and function of ACSL4, as well as the role of ACSL4 in ferroptosis. We also summarize the latest research progress of ACSL4-mediated ferroptosis in central nervous system injuries and diseases, further proving that ACSL4-medicated ferroptosis is an important target for intervention in these diseases or injuries.
Collapse
Affiliation(s)
- Bowen Jia
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Jing Li
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Yiting Song
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| | - Chengliang Luo
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University, Suzhou 215123, China
| |
Collapse
|
16
|
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
|
17
|
Lauzier DC, Jayaraman K, Yuan JY, Diwan D, Vellimana AK, Osbun J, Chatterjee AR, Athiraman U, Dhar R, Zipfel GJ. Early Brain Injury After Subarachnoid Hemorrhage: Incidence and Mechanisms. Stroke 2023; 54:1426-1440. [PMID: 36866673 PMCID: PMC10243167 DOI: 10.1161/strokeaha.122.040072] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Aneurysmal subarachnoid hemorrhage is a devastating condition causing significant morbidity and mortality. While outcomes from subarachnoid hemorrhage have improved in recent years, there continues to be significant interest in identifying therapeutic targets for this disease. In particular, there has been a shift in emphasis toward secondary brain injury that develops in the first 72 hours after subarachnoid hemorrhage. This time period of interest is referred to as the early brain injury period and comprises processes including microcirculatory dysfunction, blood-brain-barrier breakdown, neuroinflammation, cerebral edema, oxidative cascades, and neuronal death. Advances in our understanding of the mechanisms defining the early brain injury period have been accompanied by improved imaging and nonimaging biomarkers for identifying early brain injury, leading to the recognition of an elevated clinical incidence of early brain injury compared with prior estimates. With the frequency, impact, and mechanisms of early brain injury better defined, there is a need to review the literature in this area to guide preclinical and clinical study.
Collapse
Affiliation(s)
- David C. Lauzier
- Department of Neurological Surgery, Washington University School of Medicine
| | - Keshav Jayaraman
- Department of Neurological Surgery, Washington University School of Medicine
| | - Jane Y. Yuan
- Department of Neurological Surgery, Washington University School of Medicine
| | - Deepti Diwan
- Department of Neurological Surgery, Washington University School of Medicine
| | - Ananth K. Vellimana
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
- Mallinckrodt Institute of Radiology, Washington University School of Medicine
| | - Joshua Osbun
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
- Mallinckrodt Institute of Radiology, Washington University School of Medicine
| | - Arindam R. Chatterjee
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
- Mallinckrodt Institute of Radiology, Washington University School of Medicine
| | | | - Rajat Dhar
- Department of Neurology, Washington University School of Medicine
| | - Gregory J. Zipfel
- Department of Neurological Surgery, Washington University School of Medicine
- Department of Neurology, Washington University School of Medicine
| |
Collapse
|
18
|
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
|
19
|
Xie J, Lv H, Liu X, Xia Z, Li J, Hong E, Ding B, Zhang W, Chen Y. Nox4-and Tf/TfR-mediated peroxidation and iron overload exacerbate neuronal ferroptosis after intracerebral hemorrhage: Involvement of EAAT3 dysfunction. Free Radic Biol Med 2023; 199:67-80. [PMID: 36805044 DOI: 10.1016/j.freeradbiomed.2023.02.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
Intracerebral hemorrhage (ICH) induces high mortality and disability. Neuronal death is the principal factor to unfavourable prognosis in ICH. However, the mechanisms underlying this association remain unclear. In this study, we investigated the molecular mechanisms by which neuronal ferroptosis occurs after ICH and whether the use of corresponding modulators can inhibit neuronal death and improve early outcomes in a rat ICH model. Our findings indicated that Nox4 and TF/TfR were upregulated in the perihematomal tissues of ICH rats. Oxidative stress and iron overload induced by Nox4 and TF/TfR promoted neuronal ferroptosis post-ICH. In contrast, application of Nox4-siRNA and the deferoxamine (DFO) attenuated peroxidation and iron deposition in the hemorrhagic brain, alleviated neuronal ferroptosis, and improved sensorimotor function in ICH rats. Additionally, our findings indicated that the post-ICH neuronal reduced glutathione (GSH) depletion were not related to dysfunctional glutamine delivery in astrocytes but rather to downregulation of EAAT3 due to lipid peroxidation-induced dysfunction in the neuronal membrane. These findings indicate that ferroptosis is involved in neuronal death in model rats with collagenase-induced ICH. Oxidative stress and iron overload induced by Nox4 and TF/TfR exacerbate ferroptosis after ICH, while Nox4 downregulation and iron chelation exert neuroprotective effects. The present results highlight the cysteine importer EAAT3 as a potential biomarker of ferroptosis and provide insight into the neuronal death process that occurs following ICH, which may aid in the development of translational treatment strategies for ICH.
Collapse
Affiliation(s)
- Jiayu Xie
- Department of Neurosurgery, Zhujiang Hospital of Southern Medical University, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, 510282, China; Department of Neurosurgery, The First People's Hospital of Changde City of Xiangya Medical College of South Central University, Changde, 415000, China
| | - Hongzhu Lv
- Department of Neurosurgery, Zhujiang Hospital of Southern Medical University, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, 510282, China; Department of Neurosurgery, Dalian Municipal Central Hospital, Dalian, 116089, China
| | - Xuanbei Liu
- Department of Neurosurgery, Zhujiang Hospital of Southern Medical University, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, 510282, China
| | - Zhennan Xia
- Department of Neurosurgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| | - Jiangwei Li
- Department of Neurosurgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China
| | - Enhui Hong
- Department of Neurosurgery, Zhujiang Hospital of Southern Medical University, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, 510282, China
| | - Boyun Ding
- Department of Neurosurgery, Zhujiang Hospital of Southern Medical University, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, 510282, China
| | - Wenying Zhang
- Department of Neurosurgery, Zhujiang Hospital of Southern Medical University, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, 510282, China
| | - Yizhao Chen
- Department of Neurosurgery, Zhujiang Hospital of Southern Medical University, The National Key Clinical Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Engineering Technology Research Center of Education Ministry of China, Southern Medical University, Guangzhou, 510282, China; Department of Neurosurgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
20
|
Li R, Lin F, Chen Y, Lu J, Han H, Ma L, Zhao Y, Yan D, Li R, Yang J, He S, Li Z, Zhang H, Yuan K, Wang K, Hao Q, Ye X, Wang H, Li H, Zhang L, Shi G, Zhou J, Zhao Y, Zhang Y, Li Y, Wang S, Chen X, Zhao Y. A 90-Day Prognostic Model Based on the Early Brain Injury Indicators after Aneurysmal Subarachnoid Hemorrhage: the TAPS Score. Transl Stroke Res 2023; 14:200-210. [PMID: 35567655 DOI: 10.1007/s12975-022-01033-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 04/17/2022] [Accepted: 05/06/2022] [Indexed: 12/11/2022]
Abstract
This study aimed to establish a new scoring model based on the early brain injury (EBI) indicators to predict the 90-day functional outcomes in patients with aneurysmal subarachnoid hemorrhage (aSAH). We retrospectively enrolled 825 patients and prospectively enrolled 108 patients with aSAH who underwent surgical clipping or endovascular coiling (derivation cohort = 640; validation cohort = 185; prospective cohort = 108) in our institute. We established a logistic regression model based on independent risk factors associated with 90-day unfavorable outcomes. The discrimination of the prognostic model was assessed by the area under the curve in a receiver operating characteristic curve analysis. The Hosmer-Lemeshow goodness-of-fit test and a calibration plot were used to evaluate the calibration of the prediction model. The developed scoring model named "TAPS" (total score, 0-7 points) included the following admission variables: age > 55 years old, WFNS grade of 4-5, mFS grade of 3-4, Graeb score of 5-12, white blood cell count > 11.28 × 109/L, and surgical clipping. The model showed good discrimination with the area under the curve in the derivation, validation, and prospective cohorts which were 0.816 (p < 0.001, 95%CI = 0.77-0.86), 0.810 (p < 0.001, 95%CI = 0.73-0.90), and 0.803 (p < 0.001, 95%CI = 0.70-0.91), respectively. The model also demonstrated good calibration (Hosmer-Lemeshow goodness-of-fit test: X2 = 1.75, df = 8, p = 0.988). Compared with other predictive models, TAPS is an easy handle tool for predicting the 90-day unfavorable outcomes of aSAH patients, which can help clinicians better understand the concept of EBI and quickly identify those patients at risk of poor prognosis, providing more positive treatment strategies. Trial registration: NCT04785976. Registered 5 March 2021-retrospectively registered, http://www.clinicaltrials.gov .
Collapse
Affiliation(s)
- Runting Li
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Fa Lin
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Yu Chen
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Junlin Lu
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Heze Han
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Li Ma
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Yahui Zhao
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Debin Yan
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Ruinan Li
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Jun Yang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Shihao He
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Zhipeng Li
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Haibin Zhang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Kexin Yuan
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Ke Wang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Qiang Hao
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Xun Ye
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Hao Wang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
| | - Hongliang Li
- Department of Critical Care Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Linlin Zhang
- Department of Critical Care Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guangzhi Shi
- Department of Critical Care Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jianxin Zhou
- Department of Critical Care Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yang Zhao
- Department of Neurosurgery, Peking University International Hospital, Beijing, China
| | - Yukun Zhang
- Department of Neurosurgery, Peking University International Hospital, Beijing, China
| | - Youxiang Li
- Department of Interventional Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Stroke Center, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Xiaolin Chen
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China.
- Stroke Center, Beijing Institute for Brain Disorders, Beijing, China.
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.
| | - Yuanli Zhao
- Department of Neurosurgery, Fengtai District, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring West Road, Beijing, 100070, China
- Department of Neurosurgery, Peking University International Hospital, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Stroke Center, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| |
Collapse
|
21
|
Liu J, Zhou Y, Xie C, Li C, Ma L, Zhang Y. Anti-Ferroptotic Effects of bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicles Loaded with Ferrostatin-1 in Cerebral ischemia-reperfusion Injury Associate with the GPX4/COX-2 Axis. Neurochem Res 2023; 48:502-518. [PMID: 36322371 DOI: 10.1007/s11064-022-03770-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 09/13/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
Abstract
Accumulating evidence of the critical role of Ferrostatin-1 (Fer-1, ferroptosis inhibitor) in cerebral ischemia has intrigued us to explore the molecular mechanistic actions of Fer-1 delivery by bone marrow mesenchymal stem cells-derived extracellular vesicles (MSCs-EVs) in cerebral ischemia-reperfusion (I/R) injury. In vivo middle cerebral artery occlusion (MCAO) in mice and in vitro oxygen-glucose deprivation/reperfusion (OGD/R) in hippocampal neurons were developed to simulate cerebral I/R injury. After Fer-1 was confirmed to be successfully delivered by MSCs-EVs to neurons, we found that MSCs-EVs loaded with Fer-1 (MSCs-EVs/Fer-1) reduced neuron apoptosis and enhanced viability, along with curtailed inflammation and ferroptosis. The regulation of Fer-1 on GPX4/COX2 axis was predicted by bioinformatics study and validated by functional experiments. The in vivo experiments further confirmed that MSCs-EVs/Fer-1 ameliorated cerebral I/R injury in mice. Furthermore, poor expression of GPX4 and high expression of COX-2 were witnessed in cerebral I/R injury models. MSCs-EVs/Fer-1 exerted its protective effects against cerebral I/R injury by upregulating GPX4 expression and inhibiting COX-2 expression. Taken together, our study indicates that MSCs-EVs/Fer-1 may be an attractive therapeutic target for the treatment of cerebral I/R injury due to its anti-ferroptotic properties.
Collapse
Affiliation(s)
- Junying Liu
- Key Laboratory of Clinical Genetics, Affiliated Hospital & Clinical Medical College of Chengdu University, No. 82, North Section 2, 2nd Ring Road, 610081, Chengdu, Sichuan Province, P.R. China
| | - Yan Zhou
- Department of Radiation Protection Medicine, Faculty of Preventive Medicine, Air Force Medical University, 710032, Xi'an, P. R. China
| | - Chenchen Xie
- Department of Neurology, Affiliated Hospital & Clinical Medical College of Chengdu University, 610081, Chengdu, P.R. China
| | - Ci Li
- Department of Pathology, Affiliated Hospital & Clinical Medical College of Chengdu University, 610081, Chengdu, P. R. China
| | - Li Ma
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, No. 76, Huacai Road, Chenghua District, 610052, Chengdu, Sichuan Province, P. R. China.
| | - Yamei Zhang
- Key Laboratory of Clinical Genetics, Affiliated Hospital & Clinical Medical College of Chengdu University, No. 82, North Section 2, 2nd Ring Road, 610081, Chengdu, Sichuan Province, P.R. China.
| |
Collapse
|
22
|
Blocking P2RX7 Attenuates Ferroptosis in Endothelium and Reduces HG-induced Hemorrhagic Transformation After MCAO by Inhibiting ERK1/2 and P53 Signaling Pathways. Mol Neurobiol 2023; 60:460-479. [PMID: 36282438 DOI: 10.1007/s12035-022-03092-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/14/2022] [Indexed: 02/08/2023]
Abstract
Hyperglycemia is a risk factor for poor prognosis after acute ischemic stroke and promote the occurrence of hemorrhagic transformation (HT). The activation of P2RX7 play an important role in endotheliocyte damage and BBB disruption. Ferroptosis is a novel pattern of programmed cell death caused by the accumulation of intracellular iron and lipid peroxidation, resulting in ROS production and cell death. This study is to explore the mechanism of P2RX7 in reducing HT pathogenesis after acute ischemic stroke through regulating endotheliocyte ferroptosis. Male SD rats were performed to establish middle cerebral artery occlusion (MCAO) model injected with 50% high glucose (HG) and HUVECs were subjected to OGD/R treated with high glucose (30 mM) for establishing HT model in vivo and in vitro. P2RX7 inhibitor (BBG), and P2RX7 small interfering RNAs (siRNA) were used to investigate the role of P2RX7 in BBB after MCAO in vivo and OGD/R in vitro, respectively. The neurological deficits, infarct volume, degree of intracranial hemorrhage, integrity of the BBB, immunoblotting, and immunofluorescence were evaluated at 24 h after MCAO. Our study found that the level of P2RX7 was gradually increased after MCAO and/or treated with HG. Our results showed that treatment with HG after MCAO can aggravate neurological deficits, infarct volume, oxidative stress, iron accumulation, and BBB injury in HT model, and HG-induced HUVECs damage. The inhibition of P2RX7 reversed the damage effect of HG, significantly downregulated the expression level of P53, HO-1, and p-ERK1/2 and upregulated the level of SLC7A11 and GPX4, which implicated that P2RX7 inhibition could attenuate oxidative stress and ferroptosis of endothelium in vivo and in vitro. Our data provided evidence that the P2RX7 play an important role in HG-associated oxidative stress, endothelial damage, and BBB disruption, which regulates HG-induced HT by ERK1/2 and P53 signaling pathways after MCAO.
Collapse
|
23
|
Liu C, He P, Guo Y, Tian Q, Wang J, Wang G, Zhang Z, Li M. Taurine attenuates neuronal ferroptosis by regulating GABA B/AKT/GSK3β/β-catenin pathway after subarachnoid hemorrhage. Free Radic Biol Med 2022; 193:795-807. [PMID: 36402441 DOI: 10.1016/j.freeradbiomed.2022.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022]
Abstract
Ferroptosis, characterized by lipid peroxidation and intracellular iron accumulation, has been reported to be involving in the pathophysiological of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Although taurine reportedly yields neuroprotective effects in multiple central neurological diseases and can attenuated neuron damage after stroke, its role in EBI after SAH remains unclear. The present study indicated that taurine levels in cerebrospinal fluid were significantly reduced in SAH patients, which suggested that taurine treatment after SAH could improve neurological impairment, oxidative stress, iron accumulation, BBB integrity and neuronal ferroptosis in the SAH model in vivo. Taurine could attenuate MDA levels and ROS accumulation and regulate the expression of SLC7A11 and GPX4 and the AKT/GSK3β pathway in vitro. GABAB receptor inhibition and Ly294002 could reverse the therapeutic effects of taurine and significantly downregulate the levels of p-AKT, p-GSK3β, β-catenin, SLC7A11 and GPX4. The protective effects of taurine on SLC7A11 and GPX4 expression were reversed by ICG001 treatment in vitro. Taken together, our findings revealed that taurine could improve neurological function and alleviate cerebral edema, oxidative stress and BBB disruption after SAH, which reduced neuronal ferroptosis by regulating the GABAB/AKT/GSK3β/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Chengli Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Peibang He
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Yujia Guo
- 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
| | - Jianfeng Wang
- 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
| | - Zhan Zhang
- Department of Rehabilitation, 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
|
24
|
Exploring the Ferroptosis Mechanism of Zhilong Huoxue Tongyu Capsule for the Treatment of Intracerebral Hemorrhage Based on Network Pharmacology and In Vivo Validation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5033135. [PMID: 36199551 PMCID: PMC9527400 DOI: 10.1155/2022/5033135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/03/2022] [Accepted: 07/01/2022] [Indexed: 11/17/2022]
Abstract
Objective. The purpose of this study is to explore the mechanism of the Zhilong Huoxue Tongyu (ZL) capsule in the treatment of intracerebral hemorrhage (ICH) via targeting ferroptosis based on network pharmacology. Methods. The active ingredients and related key targets of the ZL capsule were screened using the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). The gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were also performed. Finally, identified targets were validated in an in-vivo model of ICH. Results. A total of 30 active ingredients and 33 intersecting targets were identified through a TCMSP database search. Ingredients-Targets-Pathways network was constructed to filter out the key targets according to the degree value. TP53 was selected as the key target. The in-vivo validation studies demonstrated that TP53 was down-regulated and GPX4 was upregulated in rats following ZL capsule treatment. Conclusions. It is concluded that the ZL capsule could alleviate ICH in a muti-target and multi-pathway manner. ZL capsule could alleviate ICH by inhibiting ferroptosis, and TP53 is identified to be the potential target. Further research is needed to clarify the detailed anti-ferroptotic mechanism of the ZL capsule.
Collapse
|
25
|
Liu Z, Zhou Z, Ai P, Zhang C, Chen J, Wang Y. Astragaloside IV attenuates ferroptosis after subarachnoid hemorrhage via Nrf2/HO-1 signaling pathway. Front Pharmacol 2022; 13:924826. [PMID: 36059982 PMCID: PMC9437486 DOI: 10.3389/fphar.2022.924826] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/13/2022] [Indexed: 11/28/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a severe type of stroke featuring exceptionally high rate of morbidity and mortality due to the lack of effective management. Ferroptosis can be defined as a novel iron-dependent programmed cell death in contrast to classical apoptosis and necrosis. Astragaloside IV (AS-IV) is an active ingredient extracted from Astragalus membranaceus with established therapeutic effect on CNS diseases. However, the exact role of ferroptosis in Astragaloside IV-mediated neuroprotection after SAH is yet to be demonstrated. In the present study, the SAH model of SD male rats with endovascular perforation was used to gauge the neuroprotective effect of AS-IV on SAH-induced early brain injury (EBI) and to clarify the potential molecular mechanism. We found that the induction of SAH reduced the levels of SLC7A11 and glutathione peroxidase 4 (GPX4) in the brain, exacerbated iron accumulation, enhanced lipid reactive oxygen species (ROS) level, and stimulated neuronal ferroptosis. However, the administration of AS-IV and the ferroptosis inhibitor Ferrostatin-1 (Fer-1) enhanced the antioxidant capacity after SAH and suppressed the accumulation of lipid peroxides. Meanwhile, AS-IV triggered Nrf2/HO-1 signaling pathway and alleviated ferroptosis due to the induction of SAH. The Nrf2 inhibitor ML385 blocked the beneficial effects of neuroprotection. These results consistently suggest that ferroptosis is profoundly implicated in facilitating EBI in SAH, and that AS-IV thwarts the process of ferroptosis in SAH by activating Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuhai Wang
- *Correspondence: Junhui Chen, ; Yuhai Wang,
| |
Collapse
|
26
|
Ren S, Chen Y, Wang L, Wu G. Neuronal ferroptosis after intracerebral hemorrhage. Front Mol Biosci 2022; 9:966478. [PMID: 35992267 PMCID: PMC9388724 DOI: 10.3389/fmolb.2022.966478] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke with high rates of morbidity, mortality, and disability. It induces cell death that is responsible for the secondary brain injury (SBI). The underlying mechanism of SBI after ICH is still unclear, and whether it is related to iron overload is worthy to be discussed. Ferroptosis is an iron-dependent non-apoptotic modes of cell death and plays a particularly important role in the occurrence and progression of ICH. Many ICH-induced regulators and signalling pathways of ferroptosis have been reported as promising targets for treating ICH. In this article, we review the definition, characteristics, and inhibition methods of neuronal ferroptosis caused by iron deposition after ICH, and review the biomarkers for ferroptosis.
Collapse
Affiliation(s)
- Siying Ren
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yue Chen
- Graduate School of Guizhou Medical University, Guiyang, China
| | - Likun Wang
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Guofeng Wu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
27
|
Tao Q, Qiu X, Li C, Zhou J, Gu L, Zhang L, Pang J, Zhang L, Yin S, Jiang Y, Peng J. S100A8 regulates autophagy-dependent ferroptosis in microglia after experimental subarachnoid hemorrhage. Exp Neurol 2022; 357:114171. [PMID: 35870523 DOI: 10.1016/j.expneurol.2022.114171] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/15/2022] [Accepted: 07/14/2022] [Indexed: 11/04/2022]
Abstract
Targeting microglial activation has been shown to ameliorate early brain injury (EBI) after subarachnoid hemorrhage (SAH). Ferroptosis is a new form of programmed cell death after SAH, but these molecular features were not recognized as evidence of microglial function so far. In this study, we constructed microglial S100A8-specific knockdown and established the SAH model in vivo and in vitro. Multi-technology strategies, including high throughput sequencing, adeno-associated virus gene gene-editing and several molecular biotechnologies to validate the effects of S100A8 on microglial autophagy and ferroptosis after SAH. Our results revealed that the expression of S100A8 was significantly increased in brain tissue after SAH. Targeted microglial S100A8 inhibition improved neural function and neuronal apoptosis in mice after SAH. Further mechanism exploration found that favourable effects of S100A8 depletion in EBI may be through the inhibition of microglia autophagy-dependent ferroptosis. In conclusion, S100A8 may be a potential intervention target for microglial ferroptosis in EBI after SAH.
Collapse
Affiliation(s)
- Qianke Tao
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiancheng Qiu
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Chaojie Li
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jian Zhou
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Long Gu
- Sichuan Clinical Research Center for Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lihan Zhang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jinwei Pang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Sichuan Clinical Research Center for Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lifang Zhang
- Sichuan Clinical Research Center for Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Shigang Yin
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China; Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Yong Jiang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China; Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| | - Jianhua Peng
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou 646000, China; Academician (Expert) Workstation of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
28
|
Liu Y, Wang Z, Cao C, Xu Z, Lu J, Shen H, Li X, Li H, Wu J, Chen G. Aquaporin 4 Depolarization-Enhanced Transferrin Infiltration Leads to Neuronal Ferroptosis after Subarachnoid Hemorrhage in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8808677. [PMID: 35761873 PMCID: PMC9233479 DOI: 10.1155/2022/8808677] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 12/19/2022]
Abstract
The infiltration of blood components into the brain parenchyma through the lymphoid system is an important cause of subarachnoid hemorrhage injury. AQP4, a water channel protein located at the astrocyte foot, has been reported to regulate blood-brain barrier integrity, and its polarization is disrupted after SAH. Neuronal ferroptosis is involved in subarachnoid hemorrhage- (SAH-) induced brain injury, but the inducing factors are not completely clear. Transferrin is one of the inducing factors of ferroptosis. This study is aimed at researching the role and mechanism of AQP4 in brain injury after subarachnoid hemorrhage in mice. An experimental mouse SAH model was established by endovascular perforation. An AAV vector encoding AQP4 with a GFAP-specific promoter was administered to mice to achieve specific overexpression of AQP4 in astrocytes. PI staining, Fer-1 intervention, and transmission electron microscopy were used to detect neuronal ferroptosis, and dextran (40 kD) leakage was used to detect BBB integrity. Western blot analysis of perfused brain tissue protein samples was used to detect transferrin infiltration. First, neuronal ferroptosis 24 h after SAH was observed by PI staining and Fer-1 intervention. Second, a significant increase in transferrin infiltration was found in the brain parenchyma 24 h after SAH modeling, while transferrin content was positively correlated with neuronal ferroptosis. Then, we observed that AQP4 overexpression effectively improved AQP depolarization and BBB injury induced by SAH and significantly reduced transferrin infiltration and neuronal ferroptosis after SAH. Finally, we found that AQP4 overexpression could effectively improve the neurobehavioral ability of SAH mice, and the neurobehavioral ability was negatively correlated with transferrin brain content. Taken together, these data indicate that overexpression of AQP4 in the mouse brain can effectively improve post-SAH neuronal ferroptosis and brain injury, at least partly by inhibiting transferrin infiltration into the brain parenchyma in the glymphatic system.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Zongqi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Chang Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Zhongmou Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street Suzhou Jiangsu Province 215006, China
- Institute of Stroke Research, Soochow University, China
| |
Collapse
|
29
|
Liu Y, Gu W. p53 in ferroptosis regulation: the new weapon for the old guardian. Cell Death Differ 2022; 29:895-910. [PMID: 35087226 PMCID: PMC9091200 DOI: 10.1038/s41418-022-00943-y] [Citation(s) in RCA: 321] [Impact Index Per Article: 107.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/05/2022] [Accepted: 01/14/2022] [Indexed: 02/08/2023] Open
Abstract
Although the conventional activities of p53 such as cell cycle arrest, senescence, and apoptosis are well accepted as the major checkpoints in stress responses, accumulating evidence implicates the importance of other tumor suppression mechanisms. Among these unconventional activities, an iron-dependent form of non-apoptotic cell death, termed ferroptosis, attracts great interest. Unlike apoptotic cell death, activation of p53 alone is not sufficient to induce ferroptosis directly; instead, through its metabolic targets, p53 is able to modulate the ferroptosis response in the presence of ferroptosis inducers such as GPX4 inhibitors or high levels of ROS. Here, we review the role of ferroptosis in p53-mediated tumor suppression, with a focus on what cellular factors are critical for p53-dependent ferroptosis during tumor suppression and how p53 modulates both the canonical (GPX4-dependent) and the non-canonical (GPX4-independent) ferroptosis pathways. We also discuss the possibility of targeting p53-mediated ferroptotic responses for the treatment of human cancers and potentially, other diseases.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
| |
Collapse
|
30
|
Ou M, Jiang Y, Ji Y, Zhou Q, Du Z, Zhu H, Zhou Z. Role and Mechanism of Ferroptosis in Neurological Diseases. Mol Metab 2022; 61:101502. [PMID: 35447365 PMCID: PMC9170779 DOI: 10.1016/j.molmet.2022.101502] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 02/08/2023] Open
Abstract
Background Ferroptosis, as a new form of cell death, is different from other cell deaths such as autophagy or senescence. Ferroptosis involves in the pathophysiological progress of several diseases, including cancers, cardiovascular diseases, nervous system diseases, and kidney damage. Since oxidative stress and iron deposition are the broad pathological features of neurological diseases, the role of ferroptosis in neurological diseases has been widely explored. Scope of review Ferroptosis is mainly characterized by changes in iron homeostasis, iron-dependent lipid peroxidation, and glutamate toxicity accumulation, of which can be specifically reversed by ferroptosis inducers or inhibitors. The ferroptosis is mainly regulated by the metabolism of iron, lipids and amino acids through System Xc−, voltage-dependent anion channels, p53, p62-Keap1-Nrf2, mevalonate and other pathways. This review also focus on the regulatory pathways of ferroptosis and its research progress in neurological diseases. Major conclusions The current researches of ferroptosis in neurological diseases mostly focus on the key pathways of ferroptosis. At the same time, ferroptosis was found playing a bidirectional regulation role in neurological diseases. Therefore, the specific regulatory mechanisms of ferroptosis in neurological diseases still need to be further explored to provide new perspectives for the application of ferroptosis in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Mengmeng Ou
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Ying Jiang
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Yingying Ji
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Qin Zhou
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Zhiqiang Du
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Haohao Zhu
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China.
| | - Zhenhe Zhou
- The affiliated Wuxi Mental Health Center of JiangNan University, Wuxi Tongren International Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China.
| |
Collapse
|
31
|
Hanke N, Rami A. Cerebral ischemia induced iron deposit, ferritin accumulation, nuclear receptor coactivator 4-depletion and ferroptosis. Curr Neurovasc Res 2022; 19:47-60. [PMID: 35319371 DOI: 10.2174/1567202619666220321120954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/04/2022] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The neuronal death upon cerebral ischemia shares not only characteristics of necrosis, apoptosis and autophagy, but exhibits also biochemical and morphological characteristics of ferroptosis. Ferroptosis is a regulated form of cell death which is considered to be an oxidative iron-dependent process. It is now commonly accepted that iron and free radicals are considered to cause lipid peroxidation as well as the oxidation of proteins and nucleic acids, leading to increased membrane and enzymatic dysfunction, and finally contributing to cell death. Although ferroptosis was first described in cancer cells, emerging evidence now links mechanisms of ferroptosis to many different diseases, including cerebral ischemia. METHODS The objective of this study was to identify the ferroptosis key players and the underlying biochemical pathways leading to cell death upon focal cerebral ischemia in mice by using immunofluorescence, Western blotting, histochemistry and densitometry. RESULTS In this study, we demonstrated that cerebral ischemia induced iron-deposition, down-regulated dramatically the expression of the glutathione peroxidase 4 (GPX4), decreased the expression of the nuclear receptor coactivator 4 (NCOA4) and induced inappropriate accumulation of ferritin in the ischemic brain. This supports the hypothesis that an ischemic insult may induce ferroptosis through inhibition of GPX4. CONCLUSION We conclude that iron excess following cerebral ischemia leads to cell death despite activation of compensatory mechanisms for iron homeostasis, as illustrated by the accumulation of ferritins. These data emphasize the presence of a cellular mechanism that allows neuronal cells to handle restriction in iron overload.
Collapse
Affiliation(s)
- Nora Hanke
- Institut für Experimentelle Neurobiologie (Anatomie II), Klinikum der Johann Wolfgang von Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| | - Abdelhaq Rami
- Institut für Experimentelle Neurobiologie (Anatomie II), Klinikum der Johann Wolfgang von Goethe-Universität, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| |
Collapse
|
32
|
Hepcidin Promoted Ferroptosis through Iron Metabolism which Is Associated with DMT1 Signaling Activation in Early Brain Injury following Subarachnoid Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:9800794. [PMID: 34987706 PMCID: PMC8723883 DOI: 10.1155/2021/9800794] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 12/28/2022]
Abstract
Iron metabolism disturbances play an important role in early brain injury (EBI) after subarachnoid hemorrhage (SAH), and hepcidin largely influences iron metabolism. Importantly, iron metabolism may be associated with ferroptosis, recently a nonapoptotic iron-dependent form of cell death that may have a great impact on brain injury after SAH. We investigated hepcidin on iron metabolism and ferroptosis involving divalent metal transporter 1 (DMT1), and ferroportin-1 (FPN1) in a rat model of SAH. Male Sprague-Dawley rats were subjected to the endovascular perforation to induce SAH, and treated with heparin (inhibitor of hepcidin), or oncostatin M (OSM, inducer of hepcidin), or ebselen (inhibitor of DMT1) by intracerebroventricular injections. Hepcidin, DMT1, FPN1 and glutathione peroxidase 4 (GPX4), were detected by western blot and immunofluorescence. Iron metabolism was detected through Perl's iron staining and iron content assay. Ferroptosis, the ROS production, lipid peroxidation (LPO) was evaluated by monitoring methane dicarboxylic aldehyde (MDA), glutathione (GSH), glutathione peroxidase 4 (GPX4) activity, and transmission electron microscopy. Neurological deficit scores, Evans blue staining and brain water content were also determined to detect EBI 72 h after SAH. Our results showed that inhibition of DMT1 by ebselen could suppress iron accumulation and lipid peroxidation, and thereby alleviate ferroptosis and EBI in SAH rats. Heparin downregulated the expression of hepcidin and DMT1, increased FPN1, and exerted protective effects that were equivalent to those of ebselen on ferroptosis and EBI. In addition, OSM increased the expression of hepcidin and DMT1, decreased FPN1, and aggravated ferroptosis and EBI, while the effect on ferroptosis was reversed by ebselen. Therefore, the study revealed that hepcidin could regulate iron metabolism and contribute to ferroptosis via DMT1 signaling activation in rats with EBI after SAH.
Collapse
|
33
|
Lu C, Tan C, Ouyang H, Chen Z, Yan Z, Zhang M. Ferroptosis in Intracerebral Hemorrhage: A Panoramic Perspective of the Metabolism, Mechanism and Theranostics. Aging Dis 2022; 13:1348-1364. [PMID: 36186133 PMCID: PMC9466971 DOI: 10.14336/ad.2022.01302] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/30/2022] [Indexed: 11/22/2022] Open
Abstract
Iron is one of the most crucial elements in the human body. In recent years, a kind of programmed, non-apoptotic cell death closely related to iron metabolism-called ferroptosis- has aroused much interest among many scientists. Ferroptosis also interacts with other pathways involved in cell death including iron abnormality, the cystine/glutamate antiporter and lipid peroxidation. Together these pathological pathways exert great impacts on intracerebral hemorrhage (ICH), a lethal cerebrovascular disease with a high incidence rate and mortality rate. Furthermore, the ferroptosis also affects different brain cells (neurons and neuroglial cells) and different organelles (mitochondria and endoplasmic reticulum). Clinical treatments for ferroptosis in ICH have been closely investigated recently. This perspective provides a comprehensive summary of ferroptosis mechanisms after ICH and its interaction with other cell death patterns. Understanding the role of ferroptosis in ICH will open new windows for the future treatments and preventions for ICH and other intracerebral diseases.
Collapse
Affiliation(s)
- Chenxiao Lu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya School of Medicine, Central South University, Changsha, 410031, China
| | - Changwu Tan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya School of Medicine, Central South University, Changsha, 410031, China
| | - Hongfei Ouyang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya School of Medicine, Central South University, Changsha, 410031, China
| | - Zhuohui Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
| | - Zhouyi Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Mengqi Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Correspondence should be addressed to: Dr. Mengqi Zhang, Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, China. ..
| |
Collapse
|