1
|
Xie Y, Wang X, Liu S, He Z, Zhang H, Yu Z, Xie M, Wang W. Transforming Growth Factor β1 Protects Against Ischemic Demyelination via Regulating Microglial Lipid Metabolism Pathway. Stroke 2025; 56:1554-1568. [PMID: 40160039 DOI: 10.1161/strokeaha.124.048206] [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: 06/17/2024] [Revised: 01/13/2025] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Chronic cerebral hypoperfusion-induced white matter lesions are an important cause of vascular cognitive impairment in aging life. TGF-β1 (transforming growth factor β1) is widely recognized as a multifunctional cytokine participating in numerous pathophysiological processes in the central nervous system. In this study, we aimed to evaluate the neuroprotective potentials of TGF-β1 in ischemic white matter lesions. METHODS A mouse model of bilateral common carotid artery stenosis was established to imitate the ischemic white matter lesions. The agonist of the TGF-β1 pathway was continuously applied via intraperitoneal injection. The Morris water maze test and gait analysis system were used to assess the cognitive and gait disorders in modeling mice. The Luxol fast blue staining, immunofluorescence, and electron microscopy were conducted to determine the severity of demyelinating lesions, microglial activation, and dysfunction of the autophagy-lysosomal pathway in microglia. Furthermore, primary cultured microglia were exposed to extracted myelin debris and TGF-β1 in vitro to explore the underlying mechanisms. RESULTS As evaluated by behavioral tests, TGF-β1 significantly alleviated the cognitive dysfunction and gait disorder in bilateral common carotid artery stenosis-modeling mice. The demyelinating lesion and remyelination process were also found to be highly improved by activation of the TGF-β1 pathway. The results of immunostaining and electron microscopy showed that TGF-β1 could ameliorate microglial activation and the dysfunction of lipid metabolism in myelin-engulfed microglia. Mechanistically, in primary cultured microglia exposed to myelin debris, administration of TGF-β1 notably mitigated the inflammatory response and accumulation of intracellular lipid droplets via promoting the lipid droplets degradation in the autophagy-lysosomal pathway, as quantified by flow cytometry, immunostaining, Western blot, etc. Yet, the application of autophagy inhibitor 3-methyladenine significantly reversed the above anti-inflammatory effects of TGF-β1. CONCLUSIONS TGF-β1 relieved cognitive deficit, demyelinating lesions, and microglia-mediated neuroinflammation in bilateral common carotid artery stenosis modeling by reducing abnormal lipid droplet accumulation and dysfunction of the autophagy-lysosomal pathway in microglia. Clinically, staged activation of the TGF-β1 pathway may become a potential target and promising treatment for ischemic white matter lesions and vascular cognitive impairment.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Xinyue Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Shuai Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College (S.L.), Huazhong University of Science and Technology, Wuhan, China
| | - Ziyu He
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College (W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| |
Collapse
|
2
|
Cao P, Chen S, Wang H, Chen Y. Taraxasterol mediated autophagy inhibition in pancreatic encephalopathy involves its regulation on L1 cell adhesion molecule. Cytotechnology 2025; 77:72. [PMID: 40051886 PMCID: PMC11880456 DOI: 10.1007/s10616-025-00721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/27/2025] [Indexed: 03/09/2025] Open
Abstract
Pancreatic encephalopathy (PE) is a frequent complication of acute pancreatitis. This study explored the mechanism of taraxasterol (TAS) in PE treatment by inhibiting pyroptosis via L1 cell adhesion molecule (L1CAM) up-regulation. PE rat models were established and treated with TAS, NLRP3 activator, and sh-L1CAM lentivirus. Serum amylase and lipase activities and Serum, hippocampus, and amygdala IL-18 and IL-1β levels were determined by ELISA, followed by TUNEL and HE staining. Rat nerve injury was evaluated by modified Neurological Severity Score (mNSS). Spontaneous behaviors, learning, memory, and emotions in rats were separately assessed by open field, new object recognition, tail suspension, and forced swimming tests. Microstructures of hippocampal CA1 region and amygdala were observed. NLRP3 + GSDMD + cells, pyroptosis markers, L1CAM, and myelin basic protein (MBP) were detected. PE rat model displayed elevated serum amylase and lipase activities and IL-18 and IL-1β levels, increased mNSS, shortened moving distance, reduced discrimination rate, prolonged immobility time, pathological damage in hippocampal CA1 region and amygdala, increased TUNEL-positive and NLRP3 + GSDMD + cells, raised NLRP3, cleaved caspase-1, GSDMD-N, IL-1β and IL-18 levels, and reduced L1CAM and MBP levels. TAS mitigated behavioral deficits and brain injury and curbed NLRP3-mediated pyroptosis in hippocampal CA1 region and amygdala in PE rats. NLRP3 activation partly averted the beneficial impacts of TAS on PE rats. TAS suppressed nerve cell pyroptosis and facilitated myelin regeneration by up-regulating L1CAM. L1CAM silencing partially abrogated TAS's effect on behavioral deficits and brain injury in PE rats. TAS treated PE by inhibiting pyroptosis via L1CAM up-regulation. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00721-x.
Collapse
Affiliation(s)
- Peng Cao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuangxi Chen
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No.69, Chuanshan Road, Shigu District, Hengyang, 421001 Hunan Province China
| | - Huiqing Wang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yanfang Chen
- Department of Neurology, The First Affiliated Hospital, Hengyang Medical School, University of South China, No.69, Chuanshan Road, Shigu District, Hengyang, 421001 Hunan Province China
| |
Collapse
|
3
|
Huang K, Zhang Q, Wan H, Ban X, Chen X, Wan X, Lu R, He Y, Xiong K. TAK1 at the crossroads of multiple regulated cell death pathways: from molecular mechanisms to human diseases. FEBS J 2025. [DOI: 10.1111/febs.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 02/14/2025] [Indexed: 05/03/2025]
Abstract
Regulated cell death (RCD), the form of cell death that can be genetically controlled by multiple signaling pathways, plays an important role in organogenesis, tissue remodeling, and maintenance of organism homeostasis and is closely associated with various human diseases. Transforming growth factor‐beta‐activated kinase 1 (TAK1) is a member of the serine/threonine protein kinase family, which can respond to different internal and external stimuli and participate in inflammatory and immune responses. Emerging evidence suggests that TAK1 is an important regulator at the crossroad of multiple RCD pathways, including apoptosis, necroptosis, pyroptosis, and PANoptosis. The regulation of TAK1 affects disease progression through multiple signaling pathways, and therapeutic strategies targeting TAK1 have been proposed for inflammatory diseases, central nervous system diseases, and cancers. In this review, we provide an overview of the downstream signaling pathways regulated by TAK1 and its binding proteins. Their critical regulatory roles in different forms of cell death are also summarized. In addition, we discuss the potential of targeting TAK1 in the treatment of human diseases, with a specific focus on neurological disorders and cancer.
Collapse
Affiliation(s)
- Kun Huang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Xiangya School of Medicine Central South University Changsha China
| | - Qi Zhang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Department of Ophthalmology Stanford University School of Medicine Palo Alto CA USA
- Key Laboratory of Emergency and Trauma of Ministry of Education, College of Emergency and Trauma Hainan Medical University Haikou China
| | - Hao Wan
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
| | - Xiao‐Xia Ban
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
| | - Xin‐Yu Chen
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
| | - Xin‐Xing Wan
- Department of Endocrinology Third Xiangya Hospital, Central South University Changsha China
| | - Rui Lu
- Department of Molecular and Cellular Physiology Stanford University Stanford CA USA
| | - Ye He
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Changsha Aier Eye Hospital China
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science Central South University Changsha China
- Key Laboratory of Emergency and Trauma of Ministry of Education, College of Emergency and Trauma Hainan Medical University Haikou China
- Hunan Key Laboratory of Ophthalmology Changsha China
| |
Collapse
|
4
|
Meng N, Su Y, Ye Z, Xie X, Liu Y, Qin C. Single-cell transcriptomic landscape reveals the role of intermediate monocytes in aneurysmal subarachnoid hemorrhage. Front Cell Dev Biol 2024; 12:1401573. [PMID: 39318997 PMCID: PMC11420033 DOI: 10.3389/fcell.2024.1401573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Objective Neuroinflammation is associated with brain injury and poor outcomes after aneurysmal subarachnoid hemorrhage (SAH). In this study, we performed single-cell RNA sequencing (scRNA-seq) to analyze monocytes and explore the mechanisms of neuroinflammation after SAH. Methods We recruited two male patients with SAH and collected paired cerebrospinal fluid (CSF) and peripheral blood (PB) samples from each patient. Mononuclear cells from the CSF and PB samples were sequenced using 10x Genomics scRNA-seq. Additionally, scRNA-seq data for CSF from eight healthy individuals were obtained from the Gene Expression Omnibus database, serving as healthy controls (HC). We employed various R packages to comprehensively study the heterogeneity of transcriptome and phenotype of monocytes, including monocyte subset identification, function pathways, development and differentiation, and communication interaction. Results (1) A total of 17,242 cells were obtained in this study, including 7,224 cells from CSF and 10,018 cells from PB, mainly identified as monocytes, T cells, B cells, and NK cells. (2) Monocytes were divided into three subsets based on the expression of CD14 and CD16: classical monocytes (CM), intermediate monocytes (IM), and nonclassical monocytes (NCM). Differentially expressed gene modules regulated the differentiation and biological function in monocyte subsets. (3) Compared with healthy controls, both the toll-like receptor (TLR) and nod-like receptor (NLR) pathways were significantly activated and upregulated in IM from CSF after SAH. The biological processes related to neuroinflammation, such as leukocyte migration and immune response regulation, were also enriched in IM. These findings revealed that IM may play a key role in neuroinflammation by mediating the TLR and NLR pathways after SAH. Interpretation In conclusion, we establish a single-cell transcriptomic landscape of immune cells and uncover the heterogeneity of monocyte subsets in SAH. These findings offer new insights into the underlying mechanisms of neuroinflammation and therapeutic targets for SAH.
Collapse
Affiliation(s)
- Ningqin Meng
- First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Ying Su
- First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Ziming Ye
- First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xufeng Xie
- The first people’s hospital of Yulin, Guangxi, China
| | - Ying Liu
- Department of Rehabilitation, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chao Qin
- First Affiliated Hospital, Guangxi Medical University, Nanning, China
| |
Collapse
|
5
|
Zhang L, Tang Y, Huang P, Luo S, She Z, Peng H, Chen Y, Luo J, Duan W, Xiong J, Liu L, Liu L. Role of NLRP3 inflammasome in central nervous system diseases. Cell Biosci 2024; 14:75. [PMID: 38849934 PMCID: PMC11162045 DOI: 10.1186/s13578-024-01256-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
The central nervous system (CNS) is the most delicate system in human body, with the most complex structure and function. It is vulnerable to trauma, infection, neurodegeneration and autoimmune diseases, and activates the immune system. An appropriate inflammatory response contributes to defence against invading microbes, whereas an excessive inflammatory response can aggravate tissue damage. The NLRP3 inflammasome was the first one studied in the brain. Once primed and activated, it completes the assembly of inflammasome (sensor NLRP3, adaptor ASC, and effector caspase-1), leading to caspase-1 activation and increased release of downstream inflammatory cytokines, as well as to pyroptosis. Cumulative studies have confirmed that NLRP3 plays an important role in regulating innate immunity and autoimmune diseases, and its inhibitors have shown good efficacy in animal models of various inflammatory diseases. In this review, we will briefly discuss the biological characteristics of NLRP3 inflammasome, summarize the recent advances and clinical impact of the NLRP3 inflammasome in infectious, inflammatory, immune, degenerative, genetic, and vascular diseases of CNS, and discuss the potential and challenges of NLRP3 as a therapeutic target for CNS diseases.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Yufen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Senlin Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Zhou She
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Hong Peng
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Yuqiong Chen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Jinwen Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Wangxin Duan
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Jie Xiong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, China.
- Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, HuChina, 410011, China.
| |
Collapse
|
6
|
Ma Y, Liu Z, Deng L, Du J, Fan Z, Ma T, Xiong J, Xiuyun X, Gu N, Di Z, Zhang Y. FGF21 attenuates neuroinflammation following subarachnoid hemorrhage through promoting mitophagy and inhibiting the cGAS-STING pathway. J Transl Med 2024; 22:436. [PMID: 38720350 PMCID: PMC11077765 DOI: 10.1186/s12967-024-05239-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) represents a form of cerebrovascular event characterized by a notable mortality and morbidity rate. Fibroblast growth factor 21 (FGF21), a versatile hormone predominantly synthesized by the hepatic tissue, has emerged as a promising neuroprotective agent. Nevertheless, the precise impacts and underlying mechanisms of FGF21 in the context of SAH remain enigmatic. METHODS To elucidate the role of FGF21 in inhibiting the microglial cGAS-STING pathway and providing protection against SAH-induced cerebral injury, a series of cellular and molecular techniques, including western blot analysis, real-time polymerase chain reaction, immunohistochemistry, RNA sequencing, and behavioral assays, were employed. RESULTS Administration of recombinant fibroblast growth factor 21 (rFGF21) effectively mitigated neural apoptosis, improved cerebral edema, and attenuated neurological impairments post-SAH. Transcriptomic analysis revealed that SAH triggered the upregulation of numerous genes linked to innate immunity, particularly those involved in the type I interferon (IFN-I) pathway and microglial function, which were notably suppressed upon adjunctive rFGF21 treatment. Mechanistically, rFGF21 intervention facilitated mitophagy in an AMP-activated protein kinase (AMPK)-dependent manner, thereby preventing mitochondrial DNA (mtDNA) release into the cytoplasm and dampening the activation of the DNA-sensing cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. Conditional knockout of STING in microglia markedly ameliorated the inflammatory response and mitigated secondary brain injuries post-SAH. CONCLUSION Our results present the initial evidence that FGF21 confers a protective effect against neuroinflammation-associated brain damage subsequent to SAH. Mechanistically, we have elucidated a novel pathway by which FGF21 exerts this neuroprotection through inhibition of the cGAS-STING signaling cascade.
Collapse
Affiliation(s)
- Yue Ma
- Department of Neurology, The affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, 710032, Shaanxi, China
| | - Zhiqin Liu
- Department of Neurology, The affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, 710032, Shaanxi, China
| | - Lele Deng
- Department of Scientific Research Section, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Jingjing Du
- Department of Neurology, The affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, 710032, Shaanxi, China
| | - Zenghui Fan
- Department of Scientific Research Section, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Tian Ma
- Department of Scientific Research Section, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Jing Xiong
- Department of Neurology, The affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, 710032, Shaanxi, China
| | - Xue Xiuyun
- Department of Neurology, The affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, 710032, Shaanxi, China
| | - Naibing Gu
- Department of Neurology, The affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, 710032, Shaanxi, China
| | - Zhengli Di
- Department of Neurology, The affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, 710032, Shaanxi, China.
| | - Yu Zhang
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
7
|
Zhao J, Chen C, Ge L, Jiang Z, Hu Z, Yin L. TAK1 inhibition mitigates intracerebral hemorrhage-induced brain injury through reduction of oxidative stress and neuronal pyroptosis via the NRF2 signaling pathway. Front Immunol 2024; 15:1386780. [PMID: 38756773 PMCID: PMC11096530 DOI: 10.3389/fimmu.2024.1386780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction Intracerebral hemorrhage (ICH) often triggers oxidative stress through reactive oxygen species (ROS). Transforming growth factor-β-activated kinase 1 (TAK1) plays a pivotal role in regulating oxidative stress and inflammation across various diseases. 5Z-7-Oxozeaenol (OZ), a specific inhibitor of TAK1, has exhibited therapeutic effects in various conditions. However, the impact of OZ following ICH and its underlying molecular mechanisms remain elusive. This study aimed to explore the possible role of OZ in ICH and its underlying mechanisms by inhibiting oxidative stress-mediated pyroptosis. Methods Adult male Sprague-Dawley rats were subjected to an ICH model, followed by treatment with OZ. Neurobehavioral function, blood-brain barrier integrity, neuronal pyroptosis, and oxidative stress markers were assessed using various techniques including behavioral tests, immunofluorescence staining, western blotting, transmission electron microscopy, and biochemical assays. Results Our study revealed that OZ administration significantly inhibited phosphorylated TAK1 expression post-ICH. Furthermore, TAK1 blockade by OZ attenuated blood-brain barrier (BBB) disruption, neuroinflammation, and oxidative damage while enhancing neurobehavioral function. Mechanistically, OZ administration markedly reduced ROS production and oxidative stress by facilitating nuclear factor-erythroid 2-related factor 2 (NRF2) nuclear translocation. This was accompanied by a subsequent suppression of the NOD-like receptor protein 3 (NLRP3) activation-mediated inflammatory cascade and neuronal pyroptosis. Discussion Our findings highlight that OZ alleviates brain injury and oxidative stress-mediated pyroptosis via the NRF2 pathway. Inhibition of TAK1 emerges as a promising approach for managing ICH.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Chunli Chen
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Lite Ge
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Jiang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhiping Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Lihong Yin
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
- Clinical Medical Research Center for Stroke Prevention and Treatment of Hunan Province, Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
8
|
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
|