1
|
Sun Z, Wang L, Ren S, Wang L, Wu G. Exploring OLR1-mediated inflammatory mechanisms in the hematoma microenvironment of acute intracerebral hemorrhage. Neuroscience 2025; 573:167-182. [PMID: 40113072 DOI: 10.1016/j.neuroscience.2025.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/09/2024] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke with high mortality and limited therapeutic options. The current study investigates the role of oxidative low-density lipoprotein receptor 1 (OLR1) within the hematoma microenvironment, focusing on its impact on immune responses and disease progression in ICH patients. Through comprehensive bioinformatics analyses of datasets from the Gene Expression Omnibus (GEO), including peripheral blood, brain tissue, and hematoma samples, we identified significant upregulation of OLR1, particularly in hematoma regions. This upregulation was strongly correlated with increased monocyte and macrophage activity, exacerbating neuroinflammation and contributing to poor clinical outcomes. Single-cell RNA sequencing (scRNA-seq) further elucidated the involvement of OLR1 in monocyte-driven immune responses, suggesting its critical role in the pathophysiology of ICH. Validation through quantitative real-time PCR (qRT-PCR) confirmed that OLR1 expression was significantly higher in hematoma samples than in peripheral blood, with the most notable elevation observed in patients with poor prognoses. Our findings suggest that OLR1 could serve as a promising biomarker and therapeutic target for modulating immune responses in ICH. Targeted therapies to regulate OLR1 expression could potentially mitigate neuroinflammation and improve recovery outcomes. This study not only enhances the understanding of the molecular mechanisms underlying ICH but also provides a foundation for developing novel therapeutic strategies that focus on the hematoma microenvironment and immune modulation.
Collapse
Affiliation(s)
- Zhilu Sun
- Clinical College, Guizhou Medical University, Guiyang 550004 Guizhou, China; Department of Emergency, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001 Hunan, China
| | - Likun Wang
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004 Guizhou, China
| | - Siying Ren
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004 Guizhou, China
| | - Long Wang
- Clinical College, Guizhou Medical University, Guiyang 550004 Guizhou, China
| | - Guofeng Wu
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004 Guizhou, China.
| |
Collapse
|
2
|
Rinald JH, Troy CM. A review of cell death pathways in hemorrhagic stroke. Front Cell Dev Biol 2025; 13:1570569. [PMID: 40356598 PMCID: PMC12066518 DOI: 10.3389/fcell.2025.1570569] [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: 02/03/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Hemorrhagic stroke is a debilitating neurological disease, affecting millions worldwide. Characterized by bleeding in the brain, it is caused by a breakdown of the blood-brain barrier (BBB) and causes damage through the presence of iron in the brain, immune activation and increased intracranial pressure. The goal of this mini-review is to explore the signaling pathways that lead to cell death that are a part of disease progression in hemorrhagic stroke. This mini-review will highlight clinical observations and data, while also incorporating findings using preclinical disease models. There are important roles for apoptosis, necroptosis, necrosis, autophagy, ferroptosis, and pyroptosis in hemorrhagic stroke. Recent work has highlighted the interplay between these phenomena, providing key regulators as potential therapeutic targets, including reactive oxygen species, iron metabolism, and caspases. Therapeutic strategies that can delay or counteract the cytotoxic effects of hemorrhage can improve clinical outcomes in hemorrhagic stroke patients.
Collapse
Affiliation(s)
- John H. Rinald
- Neurobiology and Behavior PhD Program, Columbia University, New York, NY, United States
| | - Carol M. Troy
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
3
|
Uscamaita K, Parra Ordaz O, Yazbeck Morell I, Pla MG, Sánchez-López MJ, Arboix A. From Molecular to Clinical Implications of Sleep-Related Breathing Disorders on the Treatment and Recovery of Acute Stroke: A Scoping Review. Curr Issues Mol Biol 2025; 47:138. [PMID: 40136392 PMCID: PMC11941448 DOI: 10.3390/cimb47030138] [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: 12/26/2024] [Revised: 01/28/2025] [Accepted: 02/12/2025] [Indexed: 03/27/2025] Open
Abstract
(1) Background: The aim of this review is to map research into the molecular mechanisms linking sleep-related breathing disorders (SRBDs) and acute stroke and their clinical and therapeutic implications and to identify existing knowledge gaps to suggest new areas of research. (2) Methods: This review was conducted according to the PRISMA extension for scoping reviews (PRISMA-ScR) and a predetermined protocol shared among all authors. (3) Results: The review of the thirteen studies analyzed provides a focused view of the molecular features about interaction between obstructive sleep apnea (OSA) and acute stroke. Our review identifies and highlights the biomarkers most frequently found to be associated with acute stroke, SRBDs, and their clinical repercussions. (4) Conclusions: The association between the presence of sleep apnea, especially in its severe form, and elevated levels of inflammatory markers in patients with acute stroke is highlighted and new research topics in this area are proposed.
Collapse
Affiliation(s)
- Karol Uscamaita
- Neurology Service, Sleep Disorders Unit, Hospital Universitari Sagrat Cor, Grupo Quirónsalud, 08029 Barcelona, Spain;
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain;
| | - Olga Parra Ordaz
- Medicine Department, University of Barcelona, 08036 Barcelona, Spain;
- Department of Pneumology, Sleep Disorders Unit, Hospital Universitari Sagrat Cor, Grupo Quirónsalud, 08029 Barcelona, Spain
| | - Imán Yazbeck Morell
- Department of Internal Medicine, Hospital Universitari Sagrat Cor, Grupo Quirónsalud, 08029 Barcelona, Spain;
| | - Marta García Pla
- Emergency Department, Hospital Universitari Sagrat Cor, Grupo Quirónsalud, 08029 Barcelona, Spain;
| | | | - Adrià Arboix
- Cerebrovascular Division, Department of Neurology, Hospital Universitari Sagrat Cor, Grupo Quirónsalud, Universitat de Barcelona, 08029 Barcelona, Spain
| |
Collapse
|
4
|
Wang M, Chen X, Li S, Wang L, Tang H, Pu Y, Zhang D, Fang B, Bai X. A crosstalk between autophagy and apoptosis in intracerebral hemorrhage. Front Cell Neurosci 2024; 18:1445919. [PMID: 39650799 PMCID: PMC11622039 DOI: 10.3389/fncel.2024.1445919] [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: 06/08/2024] [Accepted: 10/31/2024] [Indexed: 12/11/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is a severe condition that devastatingly harms human health and poses a financial burden on families and society. Bcl-2 Associated X-protein (Bax) and B-cell lymphoma 2 (Bcl-2) are two classic apoptotic markers post-ICH. Beclin 1 offers a competitive architecture with that of Bax, both playing a vital role in autophagy. However, the interaction between Beclin 1 and Bcl-2/Bax has not been conjunctively analyzed. This review aims to examine the crosstalk between autophagy and apoptosis in ICH by focusing on the interaction and balance of Beclin 1, Bax, and Bcl-2. We also explored the therapeutic potential of Western conventional medicine and traditional Chinese medicine (TCM) in ICH via controlling the crosstalk between autophagy and apoptosis.
Collapse
Affiliation(s)
- Moyan Wang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xin Chen
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Shuangyang Li
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Lingxue Wang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hongmei Tang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yuting Pu
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Dechou Zhang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Bangjiang Fang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Department of Emergency, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue Bai
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
5
|
Li X, Ma K, Tian T, Pang H, Liu T, Li M, Li J, Luo Z, Hu H, Hou S, Yu J, Hou Q, Song X, Zhao C, Du H, Li J, Du Z, Jin M. Methylmercury induces inflammatory response and autophagy in microglia through the activation of NLRP3 inflammasome. ENVIRONMENT INTERNATIONAL 2024; 186:108631. [PMID: 38588609 DOI: 10.1016/j.envint.2024.108631] [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: 03/19/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Methylmercury (MeHg) is a global environmental pollutant with neurotoxicity, which can easily crosses the blood-brain barrier and cause irreversible damage to the human central nervous system (CNS). CNS inflammation and autophagy are known to be involved in the pathology of neurodegenerative diseases. Meanwhile, MeHg has the potential to induce microglia-mediated neuroinflammation as well as autophagy. This study aims to further explore the exact molecular mechanism of MeHg neurotoxicity. We conducted in vitro studies using BV2 microglial cell from the central nervous system of mice. The role of inflammation and autophagy in the damage of BV2 cells induced by MeHg was determined by detecting cell viability, cell morphology and structure, reactive oxygen species (ROS), antioxidant function, inflammatory factors, autophagosomes, inflammation and autophagy-related proteins. We further investigated the relationship between the inflammatory response and autophagy induced by MeHg by inhibiting them separately. The results indicated that MeHg could invade cells, change cell structure, activate NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome and autophagosome, release a large amount of inflammatory factors and trigger the inflammatory response and autophagy. It was also found that MeHg could disrupt the antioxidant function of cells. In addition, the inhibition of NLRP3 inflammasome alleviated both cellular inflammation and autophagy, while inhibition of autophagy increased cellular inflammation. Our current research suggests that MeHg might induce BV2 cytotoxicity through inflammatory response and autophagy, which may be mediated by the NLRP3 inflammasome activated by oxidative stress.
Collapse
Affiliation(s)
- Xinyue Li
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Kai Ma
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Tiantian Tian
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Huan Pang
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Tianxiang Liu
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Meng Li
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Jiali Li
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Zhixuan Luo
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Huiyuan Hu
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Shanshan Hou
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Jing Yu
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Qiaohong Hou
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Xiuling Song
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Chao Zhao
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Haiying Du
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China
| | - Jinhua Li
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China.
| | - Zhongjun Du
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250062, PR China.
| | - Minghua Jin
- School of Public Health Jilin University, Changchun, Jilin,130021, PR China.
| |
Collapse
|
6
|
Yan D, Shi Y, Nan C, Jin Q, Zhuo Y, Huo H, Kong S, Zhao Z. Exosomes derived from human umbilical cord mesenchymal stem cells pretreated by monosialoteterahexosyl ganglioside alleviate intracerebral hemorrhage by down-regulating autophagy. Exp Cell Res 2024; 436:113960. [PMID: 38311048 DOI: 10.1016/j.yexcr.2024.113960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/06/2024]
Abstract
PURPOSE Intracerebral hemorrhage (ICH) results in substantial morbidity, mortality, and disability. Depleting neural cells in advanced stages of ICH poses a significant challenge to recovery. The objective of our research is to investigate the potential advantages and underlying mechanism of exosomes obtained from human umbilical cord mesenchymal stem cells (hUMSCs) pretreated with monosialoteterahexosyl ganglioside (GM1) in the prevention of secondary brain injury (SBI) resulting from ICH. PATIENTS AND METHODS In vitro, hUMSCs were cultured and induced to differentiate into neuron-like cells after they were pretreated with 150 μg/mL GM1. The exosomes extracted from the culture medium following a 6-h pretreatment with 150 μg/mL GM1 were used as the treatment group. Striatal infusion of collagenase and hemoglobin (Hemin) was used to establish in vivo and in vitro models of ICH. RESULTS After being exposed to 150 μg/mL GM1 for 6 h, specific cells displayed typical neuron-like cell morphology and expressed neuron-specific enolase (NSE). The rate of differentiation into neuron-like cells was up to (15.9 ± 5.8) %, and the synthesis of N-Acetylgalactosaminyltransferase (GalNAcT), which is upstream of GM1, was detected by Western blot. This study presented an increase in the synthesis of GalNAcT. Compared with the ICH group, apoptosis in the treatment group was remarkably reduced, as detected by TUNEL, and mitochondrial membrane potential was restored by JC-1. Additionally, Western blot revealed the restoration of up-regulated autophagy markers Beclin-1 and LC3 and the down-regulation of autophagy marker p62 after ICH. CONCLUSION These findings suggest that GM1 is an effective agent to induce the differentiation of hUMSCs into neuron-like cells. GM1 can potentially increase GalNAcT production through "positive feedback", which generates more GM1 and promotes the differentiation of hUMSCs. After pretreatment with GM1, exosomes derived from hUMSCs (hUMSCs-Exos) demonstrate a neuroprotective effect by inhibiting autophagy in the ICH model. This study reveals the potential mechanism by which GM1 induces differentiation of hUMSCs into neuron-like cells and confirms the therapeutic effect of hUMSCs-Exos pretreated by GM1 (GM1-Exos) on an ICH model, potentially offering a new direction for stem cell therapy in ICH.
Collapse
Affiliation(s)
- Dongdong Yan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yunpeng Shi
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chengrui Nan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qianxu Jin
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yayu Zhuo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haoran Huo
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shiqi Kong
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai, Hebei, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
7
|
Liu Z, Huang H, Yu Y, Jia Y, Li L, Shi X, Wang F. Exploring the Potential Molecular Mechanism of the Shugan Jieyu Capsule in the Treatment of Depression through Network Pharmacology, Molecular Docking, and Molecular Dynamics Simulation. Curr Comput Aided Drug Des 2024; 20:501-517. [PMID: 37340752 DOI: 10.2174/1573409919666230619105254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 04/21/2023] [Accepted: 05/11/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND Shugan Jieyu Capsule (SJC) is a pure Chinese medicine compound prepared with Hypericum perforatum and Acanthopanacis senticosi. SJC has been approved for the clinical treatment of depression, but the mechanism of action is still unclear. OBJECTIVES Network pharmacology, molecular docking, and molecular dynamics simulation (MDS) were applied in the present study to explore the potential mechanism of SJC in the treatment of depression. METHODS TCMSP, BATMAN-TCM, and HERB databases were used, and related literature was reviewed to screen the effective active ingredients of Hypericum perforatum and Acanthopanacis senticosi. TCMSP, BATMAN-TCM, HERB, and STITCH databases were used to predict the potential targets of effective active ingredients. GeneCards database, DisGeNET database, and GEO data set were used to obtain depression targets and clarify the intersection targets of SJC and depression. STRING database and Cytoscape software were used to build a protein-protein interaction (PPI) network of intersection targets and screen the core targets. The enrichment analysis on the intersection targets was conducted. Then the receiver operator characteristic (ROC) curve was constructed to verify the core targets. The pharmacokinetic characteristics of core active ingredients were predicted by SwissADME and pkCSM. Molecular docking was performed to verify the docking activity of the core active ingredients and core targets, and molecular dynamics simulations were performed to evaluate the accuracy of the docking complex. RESULTS We obtained 15 active ingredients and 308 potential drug targets with quercetin, kaempferol, luteolin, and hyperforin as the core active ingredients. We obtained 3598 targets of depression and 193 intersection targets of SJC and depression. A total of 9 core targets (AKT1, TNF, IL6, IL1B, VEGFA, JUN, CASP3, MAPK3, PTGS2) were screened with Cytoscape 3.8.2 software. A total of 442 GO entries and 165 KEGG pathways (p <0.01) were obtained from the enrichment analysis of the intersection targets, mainly enriched in IL-17, TNF, and MAPK signaling pathways. The pharmacokinetic characteristics of the 4 core active ingredients indicated that they could play a role in SJC antidepressants with fewer side effects. Molecular docking showed that the 4 core active components could effectively bind to the 8 core targets (AKT1, TNF, IL6, IL1B, VEGFA, JUN, CASP3, MAPK3, PTGS2), which were related to depression by the ROC curve. MDS showed that the docking complex was stable. CONCLUSION SJC may treat depression by using active ingredients such as quercetin, kaempferol, luteolin, and hyperforin to regulate targets such as PTGS2 and CASP3 and signaling pathways such as IL-17, TNF, and MAPK, and participate in immune inflammation, oxidative stress, apoptosis, neurogenesis, etc.
Collapse
Affiliation(s)
- Zhiyao Liu
- Department of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- High Level Key Disciplines of Traditional Chinese Medicine Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Hailiang Huang
- Department of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- High Level Key Disciplines of Traditional Chinese Medicine Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ying Yu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuqi Jia
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lingling Li
- Department of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xin Shi
- Department of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Fangqi Wang
- Department of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
8
|
Cai Y, Yu Z, Yang X, Luo W, Hu E, Li T, Zhu W, Wang Y, Tang T, Luo J. Integrative transcriptomic and network pharmacology analysis reveals the neuroprotective role of BYHWD through enhancing autophagy by inhibiting Ctsb in intracerebral hemorrhage mice. Chin Med 2023; 18:150. [PMID: 37957754 PMCID: PMC10642062 DOI: 10.1186/s13020-023-00852-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND In this study, we aimed to combine transcriptomic and network pharmacology to explore the crucial mRNAs and specific regulatory molecules of Buyang Huanwu Decoction (BYHWD) in intracerebral hemorrhage (ICH) treatment. METHODS C57BL/6 mice were randomly divided into three groups: sham, ICH, and BYHWD. BYHWD (43.29 g/kg) was administered once a day for 7 days. An equal volume of double-distilled water was used as a control. Behavioural and histopathological experiments were conducted to confirm the neuroprotective effects of BYHWD. Brain tissues were collected for transcriptomic detection. Bioinformatics analysis were performed to illustrate the target gene functions. Network pharmacology was used to predict potential targets for BYHWD. Next, transcriptomic assays were combined with network pharmacology to identify the potential differentially expressed mRNAs. Immunofluorescence staining, real-time polymerase chain reaction, western blotting, and transmission electron microscopy were performed to elucidate the underlying mechanisms. RESULTS BYHWD intervention in ICH reduced neurological deficits. Network pharmacology analysis identified 203 potential therapeutic targets for ICH, whereas transcriptomic assay revealed 109 differentially expressed mRNAs post-ICH. Among these, cathepsin B, ATP binding cassette subfamily B member 1, toll-like receptor 4, chemokine (C-C motif) ligand 12, and baculoviral IAP repeat-containing 5 were identified as potential target mRNAs through the integration of transcriptomics and network pharmacology approaches. Bioinformatics analysis suggested that the beneficial effects of BYHWD in ICH may be associated with apoptosis, animal autophagy signal pathways, and PI3K-Akt and mTOR biological processes. Furthermore, BYHWD intervention decreased Ctsb expression levels and increased autophagy levels in ICH. CONCLUSIONS Animal experiments in combination with bioinformatics analysis confirmed that BYHWD plays a neuroprotective role in ICH by regulating Ctsb to enhance autophagy.
Collapse
Affiliation(s)
- Yiqing Cai
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Zhe Yu
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Xueping Yang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Weikang Luo
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - En Hu
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Teng Li
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Wenxin Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Tao Tang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Jiekun Luo
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Regional Center for Neurological Diseases, Xiangya Hospital, Central South University Jiangxi, Nanchang, 330000, Jiangxi, People's Republic of China.
| |
Collapse
|
9
|
Wu Y, Sun Y, Wang X, Zhu C. The Regulated Cell Death and Potential Interventions in Preterm Infants after Intracerebral Hemorrhage. Curr Neuropharmacol 2023; 21:1488-1503. [PMID: 36397619 PMCID: PMC10472811 DOI: 10.2174/1570159x21666221117155209] [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: 04/21/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/21/2022] Open
Abstract
Intracerebral hemorrhage (ICH) in preterm infants is one of the major co-morbidities of preterm birth and is associated with long-term neurodevelopmental deficits. There are currently no widely accepted treatments to prevent ICH or therapies for the neurological sequelae. With studies broadening the scope of cell death, the newly defined concept of regulated cell death has enriched our understanding of the underlying mechanisms of secondary brain injury after ICH and has suggested potential interventions in preterm infants. In this review, we will summarize the current evidence for regulated cell death pathways in preterm infants after ICH, including apoptosis, necroptosis, pyroptosis, ferroptosis, autophagy, and PANoptosis as well as several potential intervention strategies that may protect the immature brain from secondary injury after ICH through regulating regulated cell death.
Collapse
Affiliation(s)
- Yanan Wu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
| | - Yanyan Sun
- Department of Human Anatomy, School of Basic Medical Science, Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
- Centre for Perinatal Medicine and Health, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou 450052, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Zhang D, Cui Y, Zhao M, Zheng X, Li C, Wei J, Wang K, Cui J. Orexin-A exerts neuroprotective effect in experimental intracerebral hemorrhage by suppressing autophagy via OXR1-mediated ERK/mTOR signaling pathway. Front Cell Neurosci 2022; 16:1045034. [PMID: 36619670 PMCID: PMC9815810 DOI: 10.3389/fncel.2022.1045034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Background Orexin-A (OXA) is a polypeptide produced in the hypothalamus, which binds to specific receptors and exerts multiple physiological effects. Autophagy plays a vital role in early brain injury (EBI) after intracerebral hemorrhage (ICH). However, the relationship between OXA and autophagy after ICH has not been confirmed. Methods In this study, the protective role of OXA was investigated in a model of hemin-induced injury in PC12 cells and blood-injection ICH model in rats, and its potential molecular mechanism was clarified. Neurobehavioral tests, brain water content, and pathologic morphology were assessed after ICH. Cell survival rate was determined using Cell Counting Kit-8 (CCK-8), while apoptosis was detected using flow cytometry. The autophagy protein LC3 that was originally identified as microtubule-associated protein 1 light 3 was evaluated by immunohistochemistry. The ultrastructural changes of cells following ICH were observed by transmission electron microscopy. Western blotting was performed to determine the expression levels of LC3, p62/SQSTM1 (p62), phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2), total extracellular signal-regulated kinase 1/2 (t-ERK1/2), mammalian target of rapamycin (mTOR), and phosphorylated mammalian target of rapamycin (p-mTOR). Results OXA treatment significantly improved neurofunctional outcomes, reduced brain edema, and alleviated neuronal apoptosis. OXA administration upregulated p-mTOR and p62, while it downregulated p-ERK1/2 and LC3; this effect was reversed by the orexin receptor 1 (OXR1) antagonist SB-334867. Conclusions This study demonstrates that OXA suppresses autophagy via the OXR1-mediated ERK/mTOR signaling pathway to exert neuroprotective effects, and it might provide a novel therapeutic approach in patients suffering from ICH.
Collapse
Affiliation(s)
- Dexin Zhang
- Department of Surgery, Hebei Medical University, Shijiazhuang, China
| | - Ying Cui
- Department of Neurology, Tangshan Gongren Hospital, Tangshan, China
| | - Manman Zhao
- Department of Histology and Embryology, North China University of Science and Technology, Tangshan, China
| | - Xuecheng Zheng
- Department of Surgery, Hebei Medical University, Shijiazhuang, China
| | - Chunyan Li
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jingbo Wei
- Department of Histology and Embryology, North China University of Science and Technology, Tangshan, China
| | - Kaijie Wang
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China
| | - Jianzhong Cui
- Department of Surgery, Hebei Medical University, Shijiazhuang, China,Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, China,*Correspondence: Jianzhong Cui,
| |
Collapse
|
11
|
Guo H, Zhang W, Wang Z, Li Z, Zhou J, Yang Z. Dexmedetomidine post-conditioning protects blood-brain barrier integrity by modulating microglia/macrophage polarization via inhibiting NF-κB signaling pathway in intracerebral hemorrhage. Front Mol Neurosci 2022; 15:977941. [PMID: 36172260 PMCID: PMC9512049 DOI: 10.3389/fnmol.2022.977941] [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: 06/25/2022] [Accepted: 08/10/2022] [Indexed: 11/18/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is one of the most devastating forms of stroke. Dexmedetomidine (DEX) has shown certain neuroprotective roles in ICH. Nevertheless, the details concerning the underlying molecular mechanism of DEX’s protective effects still need further elucidation. Herein, a model of ICH was established. The rats were randomly divided into the sham group, the ICH group, and the ICH + DEX group. Neurological outcomes, neuronal injury, and apoptosis were evaluated. Brain water content, Evans blue extravasation, and the expression of tight junction-associated proteins were also detected to assess the blood-brain barrier (BBB) integrity. Subsequently, the microglia/macrophage polarization state and inflammatory cytokine levels were observed. To further explore the underlying mechanism, NF-κB signaling pathway-associated proteins were detected. The results showed that DEX exerted neuroprotective effects against ICH-induced neurological deficits. DEX significantly increased the numbers of the surviving neurons and ameliorated neuronal cell loss and apoptosis in ICH. The rats that received the DEX displayed a lower level of brain water content and EB extravasation, moreover, ZO-1, occludin, and claudin-5 were markedly increased by DEX. Additionally, DEX facilitated M2 microglia/macrophage polarization, the M1-associated markers were reduced by DEX, while the M2-associated identification significantly increased. We found that DEX dramatically diminished pro-inflammatory cytokines expression, simultaneously promoting anti-inflammatory cytokines expression. DEX inhibited nuclear translocation of NF-κB in ICH rats. Our data suggest that DEX post-conditioning protects BBB integrity by modulating microglia/macrophage polarization via inhibiting the NF-κB signaling pathway in ICH.
Collapse
Affiliation(s)
- Hao Guo
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China.,The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Weiwei Zhang
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China
| | - Zhi Wang
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China
| | - Zhishan Li
- Department of Anesthesiology, Shanxi Provincial People's Hospital, Affiliate of Shanxi Medical University, Taiyuan, China
| | - Jing Zhou
- Shanxi Province Academy of Traditional Chinese Medicine, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, China
| | - Zhaoyu Yang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Institute of Integrative Medicine, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Zhao X, Qiao D, Guan D, Wang K, Cui Y. Chrysophanol Ameliorates Hemin-Induced Oxidative Stress and Endoplasmic Reticulum Stress by Regulating MicroRNA-320-5p/Wnt3a Pathway in HT22 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9399658. [PMID: 35936221 PMCID: PMC9355772 DOI: 10.1155/2022/9399658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022]
Abstract
Oxidative stress, endoplasmic reticulum (ER) stress, and neuronal cell apoptosis have been considered as the main pathogenesis factors of brain injury after intracerebral hemorrhage (ICH). Chrysophanol (CHR) has been proved to have neuroprotective effects, but the role and underlying mechanisms of CHR in ICH remain unclear. HT22 cells were dealt with hemin to mimic an in vitro ICH model and then subjected to treatment with or without CHR. The cell viability, apoptosis, ER stress, and oxidative stress were evaluated by conducting the cell counting kit-8 (CCK-8), TdT-mediated dUTP nick end labeling (TUNEL) staining assays, western blot, and corresponding kit, respectively. Further, microRNA-sequencing, bioinformatic analysis, dual-luciferase reporter method, and rescue experiments were conducted to explore the molecular mechanisms of CHR alleviating hemin-induced ER in HT22 cell. Our data revealed that CHR increased cells viability, antiapoptosis, anti-ER stress, and antioxidative stress under conditions of hemin-induced HT22 cell injury. Mechanically, it was observed that Wnt3a was competitively sponged by miR-320-5p, and CHR activated β-catenin pathway by regulating miR-320-5p/Wnt3a molecular axis. Finally, results from the rescue experiment suggested that CHR inhibited hemin-induced cells apoptosis, ER stress, and oxidative stress through regulating the miR-320-5p/Wnt3a axis in HT22 cells. In conclusion, CHR prevented hemin-induced apoptosis, ER stress, and oxidative stress via inhibiting the miR-320-5p/Wnt3a/β-catenin pathway in HT22 cells. Our results certified that CHR could be served as a promising treatment for brain damage following ICH.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Pharmacy, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| | - Dongge Qiao
- Nursing Department, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| | - Dongsheng Guan
- Department of Encephalopathy, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| | - Kun Wang
- Department of Pharmacy, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| | - Yinglin Cui
- Department of Encephalopathy, Henan Province Hospital of TCM (The Second Clinical Medical College, Henan University of Traditional Chinese Medicine), Zhengzhou 450002, China
| |
Collapse
|
13
|
Abd Aziz NAW, Iezhitsa I, Agarwal R, Bakar NS, Abd Latiff A, Ismail NM. Neuroprotection by Trans-Resveratrol in Rats With Intracerebral Hemorrhage: Insights into the Role of Adenosine A1 Receptors. J Neuropathol Exp Neurol 2022; 81:596-613. [PMID: 35799401 DOI: 10.1093/jnen/nlac047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Given the neuroprotective effects of trans-resveratrol (RV), this study aimed to investigate the involvement of the adenosine A1 receptor (A1R) in RV-mediated neuroprotection in a rat intracerebral hemorrhage (ICH) model induced by intrastriatal injection of collagenase. Rats were divided into 5 groups: (1) control, (2) sham-operated, (3) ICH pretreated with vehicle, (4) ICH pretreated with RV, and (5) ICH pretreated with RV and the A1R antagonist DPCPX. At 48 hours after ICH, the rats were subjected to neurological testing. Brain tissues were assessed for neuronal density and morphological features using routine and immunohistochemical staining. Expression of tumor necrosis factor-α (TNF-α), caspase-3, and RIPK3 proteins was examined using ELISA. A1R, MAPK P38, Hsp90, TrkB, and BDNF genes were examined using RT-qPCR. RV protected against neurological deficits and neuronal depletion, restored the expression of TNF-α, CASP3, RIPK3, A1R, and Hsp90, and increased BDNF/TrkB. DPCPX abolished the effects of RV on neurological outcomes, neuronal density, CASP3, RIPK3, A1R, Hsp90, and BDNF. These data indicate that the neuroprotection by RV involves A1R and inhibits CASP3-dependent apoptosis and RIPK3-dependent necroptosis in the perihematoma region; this is likely to be mediated by crosstalk between A1R and the BDNF/TrkB pathway.
Collapse
Affiliation(s)
- Noor Azliza Wani Abd Aziz
- From the Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA, NSB, NMI); Centre of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA); School of Medicine, International Medical University, Kuala Lumpur, Malaysia (II, RA); Department of Pharmacology and Bioinformatics, Volgograd State Medical University, Volgograd, Russia (II); and Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (AAL)
| | - Igor Iezhitsa
- From the Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA, NSB, NMI); Centre of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA); School of Medicine, International Medical University, Kuala Lumpur, Malaysia (II, RA); Department of Pharmacology and Bioinformatics, Volgograd State Medical University, Volgograd, Russia (II); and Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (AAL)
| | - Renu Agarwal
- From the Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA, NSB, NMI); Centre of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA); School of Medicine, International Medical University, Kuala Lumpur, Malaysia (II, RA); Department of Pharmacology and Bioinformatics, Volgograd State Medical University, Volgograd, Russia (II); and Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (AAL)
| | - Nor Salmah Bakar
- From the Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA, NSB, NMI); Centre of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA); School of Medicine, International Medical University, Kuala Lumpur, Malaysia (II, RA); Department of Pharmacology and Bioinformatics, Volgograd State Medical University, Volgograd, Russia (II); and Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (AAL)
| | - Azian Abd Latiff
- From the Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA, NSB, NMI); Centre of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA); School of Medicine, International Medical University, Kuala Lumpur, Malaysia (II, RA); Department of Pharmacology and Bioinformatics, Volgograd State Medical University, Volgograd, Russia (II); and Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (AAL)
| | - Nafeeza Mohd Ismail
- From the Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA, NSB, NMI); Centre of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh, Malaysia (NAWAA); School of Medicine, International Medical University, Kuala Lumpur, Malaysia (II, RA); Department of Pharmacology and Bioinformatics, Volgograd State Medical University, Volgograd, Russia (II); and Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia (AAL)
| |
Collapse
|
14
|
Zhang Y, Khan S, Liu Y, Zhang R, Li H, Wu G, Tang Z, Xue M, Yong VW. Modes of Brain Cell Death Following Intracerebral Hemorrhage. Front Cell Neurosci 2022; 16:799753. [PMID: 35185473 PMCID: PMC8851202 DOI: 10.3389/fncel.2022.799753] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/04/2022] [Indexed: 12/11/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke with high rates of mortality and morbidity. It induces cell death that is responsible for neurological deficits postinjury. There are no therapies that effectively mitigate cell death to treat ICH. This review aims to summarize our knowledge of ICH-induced cell death with a focus on apoptosis and necrosis. We also discuss the involvement of ICH in recently described modes of cell death including necroptosis, pyroptosis, ferroptosis, autophagy, and parthanatos. We summarize treatment strategies to mitigate brain injury based on particular cell death pathways after ICH.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Ruiyi Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Hongmin Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Guofeng Wu
- Department of Emergency, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhouping Tang
- Department of Neurology, Affiliated Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
- *Correspondence: Mengzhou Xue,
| | - V. Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- V. Wee Yong,
| |
Collapse
|
15
|
Yoshimizu A, Kinoshita K, Ichihara Y, Kurauchi Y, Seki T, Katsuki H. Hydroxychloroquine improves motor function and affords neuroprotection without inhibition of inflammation and autophagy in mice after intracerebral hemorrhage. J Neuroimmunol 2022; 362:577786. [PMID: 34920280 DOI: 10.1016/j.jneuroim.2021.577786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/28/2021] [Accepted: 12/06/2021] [Indexed: 01/05/2023]
Abstract
We examined the effect of an immunomodulator hydroxychloroquine, also known as a Nurr1 ligand and an autophagy inhibitor, on a mouse model of intracerebral hemorrhage (ICH). Daily administration of hydroxychloroquine (100 mg/kg, i.p.) from 3 h after induction of ICH alleviated neurological deficits of mice, increased the number of surviving neurons in the hematoma and prevented fragmentation of axon structures in the internal capsule. Unexpectedly, hydroxychloroquine did not inhibit either upregulation of pro-inflammatory mediators or autophagic responses in the brain. Hence, hydroxychloroquine may produce therapeutic effects on ICH primarily via neuroprotection including preservation of the axon tract integrity.
Collapse
Affiliation(s)
- Ayaka Yoshimizu
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Keita Kinoshita
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yusei Ichihara
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Takahiro Seki
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, School of Pharmacy and Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan.
| |
Collapse
|
16
|
Liu P, Yu X, Dai X, Zou W, Yu X, Niu M, Chen Q, Teng W, Kong Y, Guan R, Liu X. Scalp Acupuncture Attenuates Brain Damage After Intracerebral Hemorrhage Through Enhanced Mitophagy and Reduced Apoptosis in Rats. Front Aging Neurosci 2022; 13:718631. [PMID: 34987374 PMCID: PMC8720963 DOI: 10.3389/fnagi.2021.718631] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
To study the effect of scalp acupuncture (SA) on the mitophagy signaling pathway in the caudate nucleus of Sprague-Dawley rats following intracerebral hemorrhage (ICH). An ICH model was established by injecting autologous arterial blood into the caudate nucleus in 200 male Sprague-Dawley rats, which were divided into five groups: sham, ICH, 3-methyladenine group (3-MA, 30 mg/kg), SA, and SA+3-MA. Animals were analyzed at 6 and 24 h as well as at 3 and 7 days. Composite neurological scale score was significantly higher in the SA group than in the ICH group. Transmission electron microscopy showed less structural damage and more autophagic vacuoles within brain in the SA group than in the ICH group. SA group showed higher levels of Beclin1, Parkin, PINK1, NIX protein, and a lower level of Caspase-9 in brain tissue. These animals consequently showed less neural cell apoptosis. Compared with the SA group, however, the neural function score and levels of mitophagy protein in the SA+3-MA group were decreased, neural cell apoptosis was increased with more severe structural damage, which suggested that 3-MA may antagonize the protective effect of SA on brain in rats with ICH. SA may mitigate the neurologic impairment after ICH by enhancing mitophagy and reducing apoptosis.
Collapse
Affiliation(s)
- Peng Liu
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinyang Yu
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China.,Clinical Key Laboratory of Integrated Traditional Chinese and Western Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaohong Dai
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Zou
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China.,Clinical Key Laboratory of Integrated Traditional Chinese and Western Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xueping Yu
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Mingming Niu
- Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Qiuxin Chen
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Teng
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Kong
- Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ruiqiao Guan
- Integrated Chinese and Western Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoying Liu
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
17
|
Yuan N, Wang X, Zhang Y, Kong L, Yuan L, Ge Y. Intervention of NF-Κb Signaling Pathway and Preventing Post-Operative Cognitive Dysfunction as Well as Neuronal Apoptosis. IRANIAN JOURNAL OF PUBLIC HEALTH 2022; 51:124-132. [PMID: 35223633 PMCID: PMC8837897 DOI: 10.18502/ijph.v51i1.8303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/04/2021] [Indexed: 11/25/2022]
Abstract
Background: The Postoperative cognitive dysfunction (POCD) model was constructed by resection of the left hepatic lobe in aged mice to determine the behavioral effects of the POCD model in aged mice and the relationship between NF-κB and POCD in apoptosis and autophagy. Provide a theoretical basis for POCD prevention and treatment. Methods: This study was carried out in Ningbo No. 6 Hospital, Zhejiang, China, from Jun 2019 to Dec 2020. The POCD model was constructed after resection of the left extrahepatic lobe in aged mice and randomly divided into 6 groups: sham operation group, operation group (normal saline control group, solvent group, YC-1 group, PDTC group and 3-MA group). Related indicators of behavioral changes, neuronal inflammatory responses, apoptosis, and autophagy were examined. Results: The escape latency of the aged mice in the surgical group was significantly prolonged at three time points compared with the control group, and the number of insertions decreased significantly. Microglia are activated and the inflammatory response is increased, whereas PDTC has an inhibitory effect. It was demonstrated that apoptosis and necrosis of neurons can be induced by the NF-κb pathway, and autophagy can be promoted, whereas autophagy occurs before apoptosis. Conclusion: Activation of NF-κb pathway in neurons after POCD causes neuronal apoptosis and autophagy, and cognitive impairment occurs. PDTC, a NF-κb pathway inhibitor, can effectively reduce neuronal apoptosis induced by secondary brain injury after POCD. Necrosis, to protect the brain tissue.
Collapse
Affiliation(s)
- Na Yuan
- Department of Anaesthesia, Ningbo NO.6 Hospital, Zhejiang 315040, China
| | - Xiuzhen Wang
- Department of Anaesthesia, Ningbo NO.6 Hospital, Zhejiang 315040, China
| | - Yu Zhang
- Department of Anaesthesia, Ningbo NO.6 Hospital, Zhejiang 315040, China
| | - Lingsi Kong
- Department of Anaesthesia, Ningbo NO.6 Hospital, Zhejiang 315040, China
| | - Liyong Yuan
- Department of Anaesthesia, Ningbo NO.6 Hospital, Zhejiang 315040, China
| | - Yeying Ge
- Department of Anaesthesia, Ningbo NO.6 Hospital, Zhejiang 315040, China
| |
Collapse
|
18
|
Ebrahim HA, El-Gamal R, Sherif RN. Intermittent Fasting Attenuates High Fat Diet-Induced Cerebellar Changes in Rats: Involvement of TNFα, autophagy and oxidative stress. Cells Tissues Organs 2021; 210:351-367. [PMID: 34551416 DOI: 10.1159/000519088] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Hasnaa Ali Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Randa El-Gamal
- Department of Medical Biochemistry, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
- Medical Experimental Research Center (MERC), Faculty of Medicine, University of Mansoura, Mansoura, Egypt
| | - Rania N Sherif
- Department of Anatomy, Faculty of Medicine, University of Mansoura, Mansoura, Egypt
- Department of Anatomy and Embryology, Faculty of Dentistry, Horus University, Damietta, Egypt
- Department of Anatomy and Embryology, Faculty of Medicine, Damietta University, Damietta, Egypt
| |
Collapse
|
19
|
Wang Z, Wang G, Wang Y, Liu Q, Li H, Xie P, Wang Z. Omp31 of Brucella Inhibits NF-κB p65 Signaling Pathway by Inducing Autophagy in BV-2 Microglia. Neurochem Res 2021; 46:3264-3272. [PMID: 34536195 DOI: 10.1007/s11064-021-03429-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 02/04/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Neurobrucellosis is a serious central nervous system (CNS) inflammatory disorder caused by Brucella, and outer membrane protein-31 (Omp31) plays an important role in Brucella infection. This study aims to determine whether Omp31 can induce autophagy in BV-2 microglia. Another goal of the study is to further examine the effect of autophagy on the nuclear transcription factor κB (NF-κB) p65 signaling pathway. We observed that Omp31 stimulated autophagy by increasing microtubule-associated protein 1 light chain 3B (LC3B-II) levels and inducing autophagosome formation at 6 h and 12 h. Concomitantly, Omp31 induced tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) expression in a time-dependent manner but reduced the expression of TNF-α at 6 h. We utilized Omp31 with or without rapamycin or 3-methyladenine (3-MA) to treat BV-2 microglia, and it demonstrated further that Omp31 induced autophagy by promoting LC3B-II, Beclin-1 proteins expression and inhibiting the p62 protein levels. Furthermore, we explored the effects of autophagy on the NF-κB p65 pathway through western blot analysis, RT-qPCR assay, enzyme-linked immunosorbent assay (ELISA) and immunofluorescence. The data suggest that Omp31 as well as rapamycin, the autophagy inducer, can decrease TNF-α levels through the inhibition of the NF-κB p65 signaling pathway. Taken together, Omp31 can function as a catalyst in both autophagy induction and NF-κB p65 signal inhibition. Furthermore, Omp31-induced autophagy may inhibit the expression of TNF-α by negatively regulating NF-κB p65 signaling pathway.
Collapse
Affiliation(s)
- Zhao Wang
- Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Guowei Wang
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yanbai Wang
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Qiang Liu
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Haining Li
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Peng Xie
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Zhenhai Wang
- Neurology Center, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China. .,Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, Yinchuan, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
20
|
Tang B, Song M, Xie X, Le D, Tu Q, Wu X, Chen M. Tumor Necrosis Factor-stimulated Gene-6 (TSG-6) Secreted by BMSCs Regulates Activated Astrocytes by Inhibiting NF-κB Signaling Pathway to Ameliorate Blood Brain Barrier Damage After Intracerebral Hemorrhage. Neurochem Res 2021; 46:2387-2402. [PMID: 34145502 DOI: 10.1007/s11064-021-03375-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 12/21/2022]
Abstract
To investigate the influence of tumor necrosis factor-stimulated gene-6 (TSG-6) secreted by bone mesenchymal stem cells (BMSCs) on blood brain barrier (BBB) after intracerebral hemorrhage (ICH) and its related mechanisms. BMSCs and astrocytes were isolated and induced by TNF-α and LPS respectively. The effect of TSG-6 secreted by BMSCs on the proliferation and apoptosis of astrocytes and inflammatory response were assessed by CCK8, flow cytometry, and ELISA respectively. Then we studied the effects of TSG-6 secreted by BMSCs through the paracrine mechanism on the integrity of BBB after ICH via NF-κB signaling pathway in vitro and in vivo. We successfully isolated BMSCs and astrocytes. After LPS treatment of astrocytes, IL-1β, IL-6, and TNF-α showed an upward trend. TSG-6 secreted by TNF-α-activated BMSCs could antagonize the inflammatory response in activated astrocytes. Through the co-culture of astrocytes and BMSCs and the ICH animal model, we found that TSG-6 regulates activated astrocytes by inhibiting the NF-κB signaling pathway and ameliorates BBB damage. Furthermore, we found that TNF-α-activated BMSCs secreted exosomes containing TSG-6 and played an anti-inflammatory effect. TSG-6 secreted by BMSCs regulates activated astrocytes by inhibiting the NF-κB signaling pathway, thereby ameliorating BBB damage.
Collapse
Affiliation(s)
- Bin Tang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Min Song
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi Province, China
| | - Xun Xie
- The Second Clinical Medical College of Nanchang University, Nanchang, Jiangxi Province, China
| | - Dongsheng Le
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Qiulin Tu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Xiang Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Min Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China.
| |
Collapse
|
21
|
Guo S, Wang X, Wang L, Cheng G, Zhang M, Xing Y, Zhao X, Liu Y, Liu J. Inflammatory injury and mitophagy of the brain in chicken exposed to Cr(VI). ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:42353-42361. [PMID: 33813707 DOI: 10.1007/s11356-021-13675-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/24/2021] [Indexed: 06/12/2023]
Abstract
The aim of this study is to determine whether Cr(VI) can induce inflammatory injury in chicken brain and influence mitophagy and related mechanisms. A total of 120 hyline brown chickens (1 day old, 20±3g) were selected and randomly divided into four groups and given different doses of Cr(VI) (0, 10, 30, and 50 mg/kg) every day at 45 days. Results showed that excessive intake of Cr(VI) led to increased tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) levels and decreased interferon-gamma (IF-γ) level. Cr(VI) increased the production of mitochondrial reactive oxygen species (ROS) in chicken brain cells, causing the decline of mitochondrial membrane potential (MMP) and formation of autophagosomes for mitophagy. In addition, Cr(VI) promoted the translocation of Parkin to the mitochondrial outer membrane, increased LC3-II protein level, and inhibited p62 and TOM20 protein expression. In conclusion, excessive Cr(VI) intake can induce inflammatory injury and mitophagy in chicken brain.
Collapse
Affiliation(s)
- Shuhua Guo
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, 271018, Shandong, China
| | - Xiaozhou Wang
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, 271018, Shandong, China
| | - Lumei Wang
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, 271018, Shandong, China
| | - Guodong Cheng
- Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai`an, 271018, Shandong, China
| | - Meihua Zhang
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, 271018, Shandong, China
| | - Yuxiao Xing
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, 271018, Shandong, China
| | - Xiaona Zhao
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, 271018, Shandong, China
| | - Yongxia Liu
- Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai`an, 271018, Shandong, China.
| | - Jianzhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an, 271018, Shandong, China.
| |
Collapse
|
22
|
Zhang Y, Rui T, Luo C, Li Q. Mdivi-1 alleviates brain damage and synaptic dysfunction after intracerebral hemorrhage in mice. Exp Brain Res 2021; 239:1581-1593. [PMID: 33754161 DOI: 10.1007/s00221-021-06089-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/15/2021] [Indexed: 11/26/2022]
Abstract
As a selective inhibitor of mitochondrial fission protein dynamin-related protein-1 (Drp1), mitochondrial division inhibitor 1 (mdivi-1) can cross the blood-brain barrier (BBB) and exert neuroprotection. However, it remains unclear whether mdivi-1 can attenuate intracerebral hemorrhage (ICH)-induced secondary brain injury. This study was undertaken to characterize the roles of mdivi-1 in short-term and long-term behavioral outcomes, along with synaptic plasticity changes in mice after ICH. The results indicated mdivi-1 reversed Drp1 translocation and the morphologic changes of mitochondria, as well as ameliorated short-term neurobehavioral deficits, the BBB disruption and brain edema remarkably. In addition, mdivi-1 could rescue ICH-induced motor and memory dysfunctions. Mdivi-1 could also prevent ICH-induced reductions in synaptic proteins (synapsin I, PSD95) and phosphorylated cAMP-response element binding (p-CREB). In vitro, mdivi-1 inhibited hemin-induced hippocampal neuron death and improved neurite outgrowth. In conclusion, we found that mdivi-1 can alleviate short-term and long-term neurological deficits, synaptic dysfunction. These findings demonstrate that mdivi-1 may be beneficial in the treatment of secondary brain injury, synaptic dysfunction and neurological outcomes caused by ICH.
Collapse
Affiliation(s)
- Yunge Zhang
- Institute of Forensic Science, Changzhou De'an Hospital, Changzhou, 213003, Jiangsu, China
| | - Tongyu Rui
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, 215123, Jiangsu, China
| | - Chengliang Luo
- Department of Forensic Medicine, Medical College of Soochow University, Suzhou, 215123, Jiangsu, China.
| | - Qianqian Li
- School of Forensic Medicine, Wannan Medical College, Wuhu, 241002, Anhui, China.
| |
Collapse
|
23
|
Mechanisms of Oxidative Stress and Therapeutic Targets following Intracerebral Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8815441. [PMID: 33688394 PMCID: PMC7920740 DOI: 10.1155/2021/8815441] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/17/2021] [Accepted: 02/10/2021] [Indexed: 12/17/2022]
Abstract
Oxidative stress (OS) is induced by the accumulation of reactive oxygen species (ROS) following intracerebral hemorrhage (ICH) and plays an important role in secondary brain injury caused by the inflammatory response, apoptosis, autophagy, and blood-brain barrier (BBB) disruption. This review summarizes the current state of knowledge regarding the pathogenic mechanisms of brain injury after ICH, markers for detecting OS, and therapeutic strategies that target OS to mitigate brain injury.
Collapse
|
24
|
Huang CY, Hu RC, Li J, Chen BB, Dai AG. α1-Antitrypsin alleviates inflammation and oxidative stress by suppressing autophagy in asthma. Cytokine 2021; 141:155454. [PMID: 33611166 DOI: 10.1016/j.cyto.2021.155454] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Asthma is considered an incurable disease, although many advances have been made in asthma treatments in recent years. Therefore, elucidating the pathological mechanisms and seeking novel and effective therapeutic strategies for asthma are urgently needed. METHODS Airway resistance was measured by whole-body plethysmography. H&E staining was used to observe the morphological changes in the lung. Oxidative stress was assessed by measuring the levels of MDA, CAT and SOD. Gene expression was analysed by western blotting and RT-qPCR. ELISA was used to analyse the concentrations of IL-4, IL-5 and IFN-γ. RESULTS In the present study, we successfully established in vivo and in vitro asthma models. OVA administration led to elevated lung resistance, cell counts in BALF, and cytokine secretion, impaired airway structure and enhanced oxidative stress and autophagy in a mouse model of asthma, while IL-13 induced inflammation, oxidative stress and autophagy in BEAS-2B cells. A1AT reduced lung resistance and cell counts in BALF and suppressed inflammation, oxidative stress and autophagy in a mouse model of asthma and IL-13-induced BEAS-2B cells. Mechanistic investigations revealed that autophagy activation compromised the protective effect of A1AT on IL-13-induced BEAS-2B cells. Further mechanistic studies revealed that A1AT alleviated inflammation and oxidative stress by inhibiting autophagy in the context of asthma. CONCLUSION We demonstrated that A1AT could alleviate inflammation and oxidative stress by suppressing autophagy in the context of asthma and thus ameliorate asthma. Our study revealed novel pathological mechanisms and provided novel potential therapeutic targets for asthma treatment.
Collapse
Affiliation(s)
- Chang-Yu Huang
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410012, Hunan Province, PR China
| | - Rui-Cheng Hu
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410012, Hunan Province, PR China
| | - Jie Li
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410012, Hunan Province, PR China
| | - Bin-Bin Chen
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, Changsha 410012, Hunan Province, PR China
| | - Ai-Guo Dai
- Hunan University of Chinese Medicine, Changsha 410208, Hunan Province, PR China.
| |
Collapse
|
25
|
Gao C, Yan Y, Chen G, Wang T, Luo C, Zhang M, Chen X, Tao L. Autophagy Activation Represses Pyroptosis through the IL-13 and JAK1/STAT1 Pathways in a Mouse Model of Moderate Traumatic Brain Injury. ACS Chem Neurosci 2020; 11:4231-4239. [PMID: 33170612 DOI: 10.1021/acschemneuro.0c00517] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The newly highlighted research into programmed cell death (PCD), autophagy dependent cell death and pyroptotic cell death, has shown that these processes are both strongly correlated with the pathological progression of traumatic brain injury (TBI). However, their cross-talk in TBI remains unclear. Here, a moderate TBI model was established to explore the relationship between autophagy and pyroptosis. Rapamycin was used to activate the process of autophagy, which was impaired in the moderate TBI model, and this treatment reversed the expression of pyroptosis associated proteins, interleukin-13 (IL-13) and the pJAK-1 pathway, which were upregulated significantly after TBI. The level of IL-13 was downregulated, and the JAK-1 pathway was blocked to reveal the molecular mechanisms by which autophagy inhibits pyroptosis; these two treatments reduced the expression levels of pyroptosis associated proteins. In addition, these three interventions reduced the formation of neuronal NLRP3, the extent of brain edema, and the degree of cortical neuron degeneration. Furthermore, the deficit in motor function post-TBI was also markedly alleviated. Collectively, our results demonstrated that autophagy activation exerts a neuroprotective effect by inhibiting pyroptotic cell death in the moderate TBI model, and the inhibitory effect was dependent on the downregulation of IL-13 and repression of the JAK-1-STAT-1 signaling pathway.
Collapse
Affiliation(s)
- Cheng Gao
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou 215213, China
| | - Ya’nan Yan
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou 215213, China
| | - Guang Chen
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou 215213, China
| | - Tao Wang
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou 215213, China
| | - Chengliang Luo
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou 215213, China
| | - Mingyang Zhang
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou 215213, China
| | - Xiping Chen
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou 215213, China
| | - Luyang Tao
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou 215213, China
| |
Collapse
|
26
|
Withers SE, Parry-Jones AR, Allan SM, Kasher PR. A Multi-Model Pipeline for Translational Intracerebral Haemorrhage Research. Transl Stroke Res 2020; 11:1229-1242. [PMID: 32632777 PMCID: PMC7575484 DOI: 10.1007/s12975-020-00830-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023]
Abstract
Apart from acute and chronic blood pressure lowering, we have no specific medications to prevent intracerebral haemorrhage (ICH) or improve outcomes once bleeding has occurred. One reason for this may be related to particular limitations associated with the current pre-clinical models of ICH, leading to a failure to translate into the clinic. It would seem that a breakdown in the 'drug development pipeline' currently exists for translational ICH research which needs to be urgently addressed. Here, we review the most commonly used pre-clinical models of ICH and discuss their advantages and disadvantages in the context of translational studies. We propose that to increase our chances of successfully identifying new therapeutics for ICH, a bi-directional, 2- or 3-pronged approach using more than one model species/system could be useful for confirming key pre-clinical observations. Furthermore, we highlight that post-mortem/ex-vivo ICH patient material is a precious and underused resource which could play an essential role in the verification of experimental results prior to consideration for further clinical investigation. Embracing multidisciplinary collaboration between pre-clinical and clinical ICH research groups will be essential to ensure the success of this type of approach in the future.
Collapse
Affiliation(s)
- Sarah E Withers
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Adrian R Parry-Jones
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Stott Lane, Salford, M6 8HD, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Paul R Kasher
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
27
|
Shao A, Lin D, Wang L, Tu S, Lenahan C, Zhang J. Oxidative Stress at the Crossroads of Aging, Stroke and Depression. Aging Dis 2020; 11:1537-1566. [PMID: 33269106 PMCID: PMC7673857 DOI: 10.14336/ad.2020.0225] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/25/2020] [Indexed: 12/17/2022] Open
Abstract
Epidemiologic studies have shown that in the aging society, a person dies from stroke every 3 minutes and 42 seconds, and vast numbers of people experience depression around the globe. The high prevalence and disability rates of stroke and depression introduce enormous challenges to public health. Accumulating evidence reveals that stroke is tightly associated with depression, and both diseases are linked to oxidative stress (OS). This review summarizes the mechanisms of OS and OS-mediated pathological processes, such as inflammation, apoptosis, and the microbial-gut-brain axis in stroke and depression. Pathological changes can lead to neuronal cell death, neurological deficits, and brain injury through DNA damage and the oxidation of lipids and proteins, which exacerbate the development of these two disorders. Additionally, aging accelerates the progression of stroke and depression by overactive OS and reduced antioxidant defenses. This review also discusses the efficacy and safety of several antioxidants and antidepressants in stroke and depression. Herein, we propose a crosstalk between OS, aging, stroke, and depression, and provide potential therapeutic strategies for the treatment of stroke and depression.
Collapse
Affiliation(s)
- Anwen Shao
- 1Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Danfeng Lin
- 2Department of Surgical Oncology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Lingling Wang
- 2Department of Surgical Oncology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China
| | - Sheng Tu
- 3State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | - Cameron Lenahan
- 4Burrell College of Osteopathic Medicine, Las Cruces, USA.,5Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jianmin Zhang
- 1Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, China.,6Brain Research Institute, Zhejiang University, Zhejiang, China.,7Collaborative Innovation Center for Brain Science, Zhejiang University, Zhejiang, China
| |
Collapse
|
28
|
Molecular Correlates of Hemorrhage and Edema Volumes Following Human Intracerebral Hemorrhage Implicate Inflammation, Autophagy, mRNA Splicing, and T Cell Receptor Signaling. Transl Stroke Res 2020; 12:754-777. [PMID: 33206327 PMCID: PMC8421315 DOI: 10.1007/s12975-020-00869-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/02/2020] [Accepted: 10/18/2020] [Indexed: 12/16/2022]
Abstract
Intracerebral hemorrhage (ICH) and perihematomal edema (PHE) volumes are major determinants of ICH outcomes as is the immune system which plays a significant role in damage and repair. Thus, we performed whole-transcriptome analyses of 18 ICH patients to delineate peripheral blood genes and networks associated with ICH volume, absolute perihematomal edema (aPHE) volume, and relative PHE (aPHE/ICH; rPHE). We found 440, 266, and 391 genes correlated with ICH and aPHE volumes and rPHE, respectively (p < 0.005, partial-correlation > |0.6|). These mainly represented inflammatory pathways including NF-κB, TREM1, and Neuroinflammation Signaling-most activated with larger volumes. Weighted Gene Co-Expression Network Analysis identified seven modules significantly correlated with these measures (p < 0.05). Most modules were enriched in neutrophil, monocyte, erythroblast, and/or T cell-specific genes. Autophagy, apoptosis, HIF-1α, inflammatory and neuroinflammatory response (including Toll-like receptors), cell adhesion (including MMP9), platelet activation, T cell receptor signaling, and mRNA splicing were represented in these modules (FDR p < 0.05). Module hub genes, potential master regulators, were enriched in neutrophil-specific genes in three modules. Hub genes included NCF2, NCF4, STX3, and CSF3R, and involved immune response, autophagy, and neutrophil chemotaxis. One module that correlated negatively with ICH volume correlated positively with rPHE. Its genes and hubs were enriched in T cell-specific genes including hubs LCK and ITK, Src family tyrosine kinases whose modulation improved outcomes and reduced BBB dysfunction following experimental ICH. This study uncovers molecular underpinnings associated with ICH and PHE volumes and pathophysiology in human ICH, where knowledge is scarce. The identified pathways and hub genes may represent novel therapeutic targets.
Collapse
|
29
|
Chen J, Wang H, Luo C, Gao C, Zhang Y, Chen G, Chen W, Chen X, Tao L. Chd8 Rescued TBI-Induced Neurological Deficits by Suppressing Apoptosis and Autophagy Via Wnt Signaling Pathway. Cell Mol Neurobiol 2020; 40:1165-1184. [PMID: 32034634 PMCID: PMC11448946 DOI: 10.1007/s10571-020-00806-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 01/30/2020] [Indexed: 12/30/2022]
Abstract
Traumatic brain injury (TBI) and autism spectrum disorder (ASDs) share several same biochemical mechanisms and symptoms, such as learning memory impairments and communication deficits. Chromodomain helicase DNA binding protein 8 (CHD8), a member of the CHD family of ATP-dependent chromatin remodeling factors, is one of the top risk genetic factors in ASDs and is highly associated with Wnt/β-catenin signaling. Yet, the possible effect of CHD8 on TBI remains poorly understood. In vivo, we found that Chd8 co-localized in neurons, astrocytes, and microglia, but predominantly presented in neurons in the prefrontal cortex, hippocampus, and cortex. Both Chd8 and β-catenin expression peaked at 12 h and shared the similar change tendency after TBI. Chd8 knockdown inhibited wnt pathway, promoted the activation of apoptosis and autophagy, and caused learning and memory impairments both at normal and TBI condition. In addition, overexpression of Chd8 via 17β-estrogen (E2) treatment enhanced wnt signaling pathway and suppressed TBI-induced apoptosis and autophagic activation. In vitro, a significant increase of Chd8 and β-catenin expression was observed in HT22 cells after lipopolysaccharide (lps) treatment or mechanical injury, respectively. Chd8 knockdown inhibited wnt signaling pathway and increased apoptosis and autophagy activation in lps-stimulated HT22 cells. But activation of wnt signaling inverted the effects of Chd8-siRNA. Our results demonstrated that Chd8 exerted neuroprotection and promoted cognitive recovery through inhibiting apoptosis and autophagy activation following TBI, at least partially by wnt signaling pathway.
Collapse
Affiliation(s)
- Jie Chen
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou, 215023, China
| | - Haochen Wang
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou, 215023, China
| | - Chengliang Luo
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou, 215023, China
| | - Cheng Gao
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou, 215023, China
| | - Yalai Zhang
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou, 215023, China
| | - Guang Chen
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou, 215023, China
| | - Wei Chen
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou, 215023, China
| | - Xiping Chen
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou, 215023, China.
| | - Luyang Tao
- Department of Forensic Medicine, Medical School of Soochow University, 178 East Ganjiang Road, Suzhou, 215023, China.
| |
Collapse
|
30
|
Yang Z, Zhou C, Shi H, Zhang N, Tang B, Ji N. Heme Induces BECN1/ATG5-Mediated Autophagic Cell Death via ER Stress in Neurons. Neurotox Res 2020; 38:1037-1048. [PMID: 32840757 DOI: 10.1007/s12640-020-00275-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/11/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022]
Abstract
Intracerebral hemorrhage (ICH) is a serious medical problem, and effective treatment is limited. Hemorrhaged blood is highly toxic to the brain, and heme, which is mainly released from hemoglobin, plays a vital role in neurotoxicity. However, the specific mechanism involved in heme-mediated neurotoxicity has not been well studied. In this study, we investigated the neurotoxicity of heme in neurons. Neurons were treated with heme, and cell death, autophagy, and endoplasmic reticulum (ER) stress were analyzed. In addition, the relationship between autophagy and apoptosis in heme-induced cell death and the downstream effects were also assessed. We showed that heme induced cell death and autophagy in neurons. The suppression of autophagy using either pharmacological inhibitors (3-methyladenine) or RNA interference of essential autophagy genes (BECN1 and ATG5) decreased heme-induced cell death in neurons. Moreover, the ER stress activator thapsigargin increased cell autophagy and the cell death ratio following heme treatment. Autophagy promoted heme-induced cell apoptosis and cell death through the BECN1/ATG5 pathway. Our findings suggest that heme potentiates neuronal autophagy via ER stress, which in turn induces cell death via the BECN1/ATG5 pathway. Targeting ER stress-mediated autophagy might be a promising therapeutic strategy for ICH.
Collapse
Affiliation(s)
- Zhao Yang
- Department of Neurology and Chongqing Key Laboratory of Cerebrovascular Disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Changlong Zhou
- Department of Neurology and Chongqing Key Laboratory of Cerebrovascular Disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Hui Shi
- Department of Neurology and Chongqing Key Laboratory of Cerebrovascular Disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Nan Zhang
- Department of Urology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Bin Tang
- Department of General Surgery, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China.
| | - Na Ji
- Department of Anesthesia, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
31
|
Xiao H, Chen H, Jiang R, Zhang L, Wang L, Gan H, Jiang N, Zhao J, Zhai X, Liang P. NLRP6 contributes to inflammation and brain injury following intracerebral haemorrhage by activating autophagy. J Mol Med (Berl) 2020; 98:1319-1331. [PMID: 32783081 DOI: 10.1007/s00109-020-01962-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022]
Abstract
Inflammation is a crucial factor contributing to secondary brain injury after intracerebral haemorrhage (ICH). NLRP6, a member of nod-like receptors (NLRs) family, has been reported to participate in inflammation and host-defence in multiple diseases. Distinct from the other NLR family members, NLRP6 regulates inflammation in an inflammasome-dependent as well as an inflammasome-independent pathway. However, the role of NLRP6 in regulating signalling pathways during ICH is poorly understood. In the present study, we demonstrated that NLRP6 expression was upregulated after ICH, both in humans and in rats. Subsequently, we developed a rat model of ICH and found that NLRP6 knockdown reduced brain injury, alleviated inflammation, and suppressed autophagy following ICH. Further, results indicated that autophagy involved in NLRP6 mediated inflammation after ICH. Moreover, we found that NLRP6 mediated regulation of autophagy and inflammation was inflammasome-dependent. This study revealed the underlying molecular mechanism of NLRP6 in inflammation and highlights the therapeutic potential of targeting NLRP6 in secondary brain injury after ICH. KEY MESSAGES: • NLRP6 was upregulated following ICH in humans and rats. • NLRP6 knockdown reduced brain injury, alleviated inflammation, and suppressed autophagy following ICH. • NLRP6 aggravated inflammation after ICH by activating autophagy. • NLRP6 regulated inflammation and autophagy after ICH by activating inflammasome pathway.
Collapse
Affiliation(s)
- Han Xiao
- Department of Neurosurgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Hui Chen
- Department of Neurosurgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Neurosurgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Lu Wang
- Department of Neurosurgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Hui Gan
- Department of Neurosurgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China.,Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Ning Jiang
- Institute of Neuroscience, Chongqing Medical University, Chongqing, China.,Department of Pathology, Chongqing Medical University, Chongqing, China
| | - Jing Zhao
- Institute of Neuroscience, Chongqing Medical University, Chongqing, China.,Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Xuan Zhai
- Department of Neurosurgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China.,Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Ping Liang
- Department of Neurosurgery, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China. .,Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China. .,Institute of Neuroscience, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
32
|
Li J, Ye M, Gao J, Zhang Y, Zhu Q, Liang W. Systematic Understanding of Mechanism of Yi-Qi-Huo-Xue Decoction Against Intracerebral Hemorrhagic Stroke Using a Network Pharmacology Approach. Med Sci Monit 2020; 26:e921849. [PMID: 32769962 PMCID: PMC7433745 DOI: 10.12659/msm.921849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH), a fatal type of stroke, profoundly affects public health. Yi-Qi-Huo-Xue decoction (YQHXD), a traditional Chinese medicine (TCM) prescription, is verified to be an efficient method to treat ICH stroke among the Chinese population. Nevertheless, the pharmacological mechanisms of YQHXD have been unclear. Material/Methods We used a strategy based on network pharmacology to explore the possible multi-component, multi-target, and multi-pathway pattern of YQHXD in treating ICH. First, candidate targets for YQHXD were identified using the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Then, these candidate YQHXD targets were used in combination with the known targets for the treatment of ICH stroke to construct the core network (cPPI) using data on protein–protein interaction (PPI). We calculated 5 topological parameters for identification of the main hubs. Pathway enrichment and GO biological process enrichment analyses were performed after the incorporation of the main hubs into ClueGO. Results In total, 55 candidate YQHXD targets for ICH were recognized to be the major hubs in accordance with their topological importance. As suggested by enrichment analysis, the YQHXD targets for ICH were roughly classified into several biological processes (related to redox equilibrium, cell–cell communication, adhesion and collagen biosynthesis, cytokine generation, lymphocyte differentiation and activation, neurocyte apoptosis and development, neuroendocrine system, and vascular development) and related pathways (VEGF, mTOR, NF-kB, RAS/MAPK, JAK/STAT and cytokine–cytokine receptors interaction), indicating those mechanisms underlying the therapeutic effect of YQHXD. Conclusions The present results may serve as a pharmacological framework for TCM studies in the future, helping to promote the use of YQHXD in clinical treatment of ICH.
Collapse
Affiliation(s)
- Jian Li
- Department of Neurosurgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Ming Ye
- Department of Neurosurgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Jueming Gao
- Department of Neurosurgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Yeqing Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Qiyong Zhu
- Department of Respiratory Medicine, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| | - Weibang Liang
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
33
|
Zhang H, Lu X, Hao Y, Tang L, He Z. MicroRNA-26a-5p alleviates neuronal apoptosis and brain injury in intracerebral hemorrhage by targeting RAN binding protein 9. Acta Histochem 2020; 122:151571. [PMID: 32622424 DOI: 10.1016/j.acthis.2020.151571] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/15/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023]
Abstract
Emerging evidence has unraveled the important implications of microRNAs (miRNAs/miRs) in intracerebral hemorrhage (ICH). The aim of the present study was to assess the possible regulatory role of miR-26a-5p in ICH both in vivo and in vitro. ICH model of rats was constructed using stereotactic injection of VII collagenase, and ICH condition of PC-12 cells was stimulated by hemin. Exogenous overexpression of miR-26a-5p was achieved utilizing the transfection with miR-26a-5p agomir or miR-26a-5p mimics. We detected decreased miR-26a-5p and increased RAN binding protein 9 (RANBP9) levels in perihematomal tissues of ICH rats and in PC-12 cells following ICH. While miR-26a-5p overexpression alleviated behavioral deficits and neuronal apoptosis of rats with ICH. Apoptosis-related proteins Bax, Bcl-2 and cleaved caspase-3 in perihematomal region were also measured to further confirm the inhibitory effect of miR-26a-5p on neuronal apoptosis. In ICH models in vitro, we found that miR-26a-5p overexpression significantly decreased hemin-stimulated apoptosis of PC-12 cells. Additionally, RANBP9 knockdown could suppress the apoptosis of PC-12 cells, similar to the effects of PC-12 cells transfected with miR-26a-5p mimics. With dual-luciferase reporter assay, we identified that miR-26a-5p directly targeted RANBP9. In conclusion, exogenous miR-26a-5p alleviated neuronal apoptosis and brain injury partially by targeting RANBP9, and miR-26a-5p/RANBP9 axis may be a potential target for ICH treatment.
Collapse
|
34
|
MST4 Kinase Inhibitor Hesperadin Attenuates Autophagy and Behavioral Disorder via the MST4/AKT Pathway in Intracerebral Hemorrhage Mice. Behav Neurol 2020; 2020:2476861. [PMID: 32089749 PMCID: PMC7023841 DOI: 10.1155/2020/2476861] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 11/14/2019] [Accepted: 01/08/2020] [Indexed: 01/14/2023] Open
Abstract
Background The aim of this study was to explore the role of hesperadin in intracerebral hemorrhage (ICH) mice, with the involvement of the mammalian ste20-like kinase 4 (MST4)/AKT signaling pathway. Methods All mice were divided into four groups: sham group, sham+hesperidin group, ICH group, and ICH+hesperadin group. The effects of hesperadin were assessed on the basis of brain edema and neurobehavioral function. Furthermore, we observed MST4, AKT, phosphorylation of AKT (pAKT), and microtubule-associated protein light chain 3 (LC3) by western blotting. Protein localization of MST4 and LC3 was determined by immunofluorescence. Results The expression of MST4 was upregulated at 12 h and 24 h after ICH. Brain edema was significantly decreased and neurological function was improved in the hesperadin treatment group compared to the ICH group (P < 0.05). Hesperadin decreases the expressions of MST and increases pAKT after ICH. Autophagy significantly increased in the ICH group, while hesperadin reduced this increase. Conclusion Hesperadin provides neuroprotection against ICH by inhibiting the MST4/AKT signaling pathway.
Collapse
|
35
|
Huan S, Jin J, Shi CX, Li T, Dai Z, Fu XJ. Overexpression of miR-146a inhibits the apoptosis of hippocampal neurons of rats with cerebral hemorrhage by regulating autophagy. Hum Exp Toxicol 2020; 39:1178-1189. [PMID: 32090627 DOI: 10.1177/0960327120907131] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In this study, to investigate the effect of overexpression of miR-146a on autophagy of hippocampal neurons in rats with intracerebral hemorrhage (ICH), 72 Sprague-Dawley rats were randomly divided into the sham, ICH, miR-146a agomir, and miR-146a agomir control groups. The ICH model was constructed by injection of collagenase VII. The apoptosis of hippocampal neurons was measured by TUNEL assay. The levels of LC3 and Beclin 1 were analyzed by immunohistochemistry. Mitochondrial autophagy was examined by transmission electron microscopy. The levels of LC3A, LC3B, Beclin 1, Bax, Bcl-2, and cleaved caspase 3 were examined by Western blot. Western blot was also used to evaluate the expression of nuclear factor κB signaling pathway-related factors. To examine the effect of autophagy inhibitor (3-methyladenine (3-MA)) on miR-146a-regulated apoptotic protein expression, 30 rats were further divided into the sham, ICH, miR-146a agomir, 3-MA, and miR-146a + 3-MA groups. The levels of Bax, Bcl-2, and cleaved caspase 3 were examined by Western blot. Compared with the sham group, the nerve function scores, brain water content, the percentage of apoptotic cells, and the expression levels of LC3, Beclin 1, Bax, cleaved caspase 3, and p-P65 in the hippocampus of rats in the ICH group were all significantly increased (p < 0.05), whereas the expression levels of miR-146a, Bcl-2, and p-IκBα were markedly decreased (p < 0.05). Mitochondrial autophagy was also evident. Furthermore, compared with the ICH group, the results of the abovementioned tests in the miR-146a agomir group were reversed. The overexpression of miR-146a inhibited the autophagy of hippocampal neurons in rats with ICH.
Collapse
Affiliation(s)
- S Huan
- Department of Rehabilitation Sciences, Qingdao Women and Children Hospital, Qingdao University, Qingdao, China
| | - J Jin
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - C-X Shi
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - T Li
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Z Dai
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - X-J Fu
- Department of Nephrology, Huai'an Hospital Affiliated to Xuzhou Medical University and Huai'an Second Hospital, Huai'an, China
| |
Collapse
|
36
|
Abd Aziz NAW, Iezhitsa I, Agarwal R, Abdul Kadir RF, Abd Latiff A, Ismail NM. Neuroprotection by trans-resveratrol against collagenase-induced neurological and neurobehavioural deficits in rats involves adenosine A1 receptors. Neurol Res 2020; 42:189-208. [PMID: 32013788 DOI: 10.1080/01616412.2020.1716470] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective: Trans-resveratrol has been shown to have neuroprotective effects and could be a promising therapeutic agent in the treatment of intracerebral haemorrhage (ICH). This study aimed to investigate the involvement of the adenosine A1 receptor (A1R) in trans-resveratrol-induced neuroprotection in rats with collagenase-induced ICH.Methods: Sixty male Sprague-Dawley rats weighing 330-380 g were randomly divided into five groups (n = 12): (i) control, (ii) sham-operated rats, (iii) ICH rats pretreated with vehicle (0.1% DMSO saline, i.c.v.), (iv) ICH rats pretreated with trans-resveratrol (0.9 µg, i.c.v.) and (v) ICH rats pretreated with trans-resveratrol (0.9 µg) and the A1R antagonist, DPCPX (2.5 µg, i.c.v.). Thirty minutes after pretreatment, ICH was induced by intrastriatal injection of collagenase (0.04 U). Forty-eight hours after ICH, the rats were assessed using a variety of neurobehavioural tests. Subsequently, rats were sacrificed and brains were subjected to gross morphological examination of the haematoma area and histological examination of the damaged area.Results: Severe neurobehavioural deficits and haematoma with diffuse oedema were observed after intrastriatal collagenase injection. Pretreatment with trans-resveratrol partially restored general locomotor activity, muscle strength and coordination, which was accompanied with reduction of haematoma volume by 73.22% (P < 0.05) and damaged area by 60.77% (P < 0.05) in comparison to the vehicle-pretreated ICH group. The trans-resveratrol-induced improvement in neurobehavioural outcomes and morphological features of brain tissues was inhibited by DPCPX pretreatment.Conclusion: This study demonstrates that the A1R activation is possibly the mechanism underlying the trans-resveratrol-induced neurological and neurobehavioural protection in rats with ICH.
Collapse
Affiliation(s)
- Noor Azliza Wani Abd Aziz
- Centre for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia.,Centre of PreClinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh, Malaysia
| | - Igor Iezhitsa
- Centre for Neuroscience Research, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia.,Research Centre for Innovative Medicines, Volgograd State Medical University, Volgograd, Russia.,Institute for Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA, Sungai Buloh, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | | | - Azian Abd Latiff
- Department of Anatomy, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh, Malaysia
| | - Nafeeza Mohd Ismail
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Hu L, Zhang H, Wang B, Ao Q, He Z. MicroRNA-152 attenuates neuroinflammation in intracerebral hemorrhage by inhibiting thioredoxin interacting protein (TXNIP)-mediated NLRP3 inflammasome activation. Int Immunopharmacol 2020; 80:106141. [PMID: 31982825 DOI: 10.1016/j.intimp.2019.106141] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/30/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022]
Abstract
Neuroinflammation significantly contributes to brain injury and neurological deterioration following intracerebral hemorrhage (ICH). MicroRNA-152(miR-152) was reported to be downregulated in ICH patients and to possess anti-inflammatory properties in other diseases. In this study, we aimed to explore the role of miR-152 in ICH, and the underlying mechanisms, using a collagenase-induced rat ICH model and hemin-exposure as a cell model. We first confirmed that miR-152 was consistently downregulated in both models. Overexpression of miR-152 in microglial BV2 cells reduced hemin-induced inflammatory response and reactive oxygen species (ROS) generation, thus protecting co-cultured neuronal HT22 cells. Moreover, overexpression of miR-152 by intracerebroventricular lentivirus injection in ICH rats significantly alleviated neurodecifits, brain edema, and hematoma. These changes were associated with a marked reduction in ICH-induced neuronal death, as detected by co-staining of NeuN and TUNEL, and ICH-induced neuroinflammation, as revealed by inflammatory cytokine levels as well as by the number of Iba1 positive-stained cells in the perihematomal region. Mechanistically, miR-152 significantly inhibited ICH-induced TXNIP expression, and its overexpression blocked the interaction between TXNIP and NOD-like receptor pyrin domain containing 3(NLRP3), thus inhibiting NLRP3-driven inflammasome activation to attenuate neuroinflammation in vivo and in vitro. Moreover, the results of si-TXNIP transfection further confirmed that TXNIP inhibition was involved in the reduction of NLRP3 inflammasome activation by the overexpression of miR-152. Collectively, the present study demonstrates that miR-152 confers protection against ICH-induced neuroinflammation and brain injury by inhibiting TXNIP-mediated NLRP3 inflammasome activation, indicating a potential strategy for ICH treatment.
Collapse
Affiliation(s)
- Liuting Hu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110000, People's Republic of China
| | - Heyu Zhang
- Department of Neurology, The First Affiliated Hospital Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Bingyang Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110000, People's Republic of China
| | - Qiang Ao
- Department of Tissue Engineering, China Medical University, Shenyang 110122, People's Republic of China
| | - Zhiyi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang 110000, People's Republic of China.
| |
Collapse
|
38
|
Inhibiting nuclear factor-κB at different stages after intracerebral hemorrhage can influence the hemorrhage-induced brain injury in experimental models in vivo. Brain Res Bull 2019; 155:159-165. [PMID: 31857135 DOI: 10.1016/j.brainresbull.2019.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/27/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Nuclear factor-κB (NF-κB) is a critical regulator of inflammatory responses after ICH, and different subunits may have different influences on the cell death and prognosis. The aim of the present study is to clarify whether the prognosis can be influenced by inhibiting NF-κB activation and subunits expression using PDTC at different stages after ICH. METHODS Rats were divided into sham group, ICH group, early interference group and late interference group. At preset time points after ICH, the ipsilateral striatum and tissue around was obtained for detection of NF-κB activation, cell death, and expression of caspase-3, bcl-2, and NF-κB subunits, to evaluate of the effect of PDTC. RESULTS NF-κB subunit p65 mainly expressed at the early stage after ICH, and c-Rel at the late stage. NF-κB activation can be inhibited at the early stage after ICH by administrating PDTC at 10 min, 1d and 2d after ICH, and at the late stage at 6d,7d and 8d. NF-κB activation inhibition at the early stage was due to p65, and c-Rel at the late stage. Inhibiting p65 expression at the early stage after ICH can reduce the apoptotic factor caspase-3 expression and cell death, and raise the antiapoptotic factor bcl-2. Meanwhile, inhibiting c-Rel expression at the late stage after ICH can lead to the opposite result. CONCLUSION Measures of inhibiting NF-κB subunits can be performed to influence the secondary brain damage and prognosis of ICH. We can also speculate that early inhibition of p65 expression and late promotion of c-Rel expression may be a more efficient method to improve the prognosis of ICH.
Collapse
|
39
|
Hu L, Zhang H, Wang B, Ao Q, Shi J, He Z. MicroRNA-23b alleviates neuroinflammation and brain injury in intracerebral hemorrhage by targeting inositol polyphosphate multikinase. Int Immunopharmacol 2019; 76:105887. [PMID: 31536904 DOI: 10.1016/j.intimp.2019.105887] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/21/2019] [Accepted: 09/05/2019] [Indexed: 12/27/2022]
Abstract
Neuroinflammation plays a critical role in the pathogenesis of intracerebral hemorrhage (ICH), contributing to detrimental brain injury and neurological function deficits. MicroRNA-23b (miR-23b) exerts anti-inflammatory effects in many diseases and is downregulated in patients with ICH. This study aimed to evaluate the involvement of miR-23b in ICH models in vivo and in vitro, using basal ganglia injection of collagenase type VII in rats and hemin stimulation for cells, respectively. Exogenous overexpression of miR-23b by transfection with lentivirus-miR-23b (LV-miR-23b) or miR-23b mimics was evaluated by RT-qPCR. In this study, we found miR-23b was downregulated in the ICH models and its overexpression effectively alleviated neurological deficits, brain edema, hematoma area, and neuronal apoptosis in ICH rats. Western blotting for neuroinflammation markers and immunofluorescence staining for microglial activation demonstrated that miR-23b could alleviate neuroinflammation in ICH in vivo. We also performed an in vitro mechanism study using BV2 microglial cells and HT22 neuronal cell lines to explore how miR-23b modulates neuroinflammation and neuronal protection after ICH. We found that miR-23b significantly decreased hemin-stimulated inflammation response in BV2 cells and attenuated co-cultured HT22 neuronal cell death. Additionally, we verified that miR-23b suppressed inflammation in BV2 cells by targeting inositol polyphosphate multikinase (IPMK) and that autophagy regulation through the Akt/mTOR pathway was involved in miR-23b-regulated inflammation after ICH. Our study illustrated that miR-23b played a protective role in ICH through inhibiting neuroinflammation by targeting IPMK; this mechanism may be related to the regulation of the Akt/mTOR autophagy pathway, making it a potential target for ICH treatment.
Collapse
Affiliation(s)
- Liuting Hu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110000,People's Republic of China
| | - Heyu Zhang
- Dapartment of Neurology, The First Affiliated Hospital Sun Yat-sen University,Guangzhou 510080,People's Republic of China
| | - Bingyang Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110000,People's Republic of China
| | - Qiang Ao
- Department of Tissue Engineering, China Medical University, Shenyang 110122, People's Republic of China
| | - Jing Shi
- Department of Neurology, Dandong Central Hospital, Dandong 118002,People's Republic of China
| | - Zhiyi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110000,People's Republic of China.
| |
Collapse
|
40
|
Shan H, Qiu J, Chang P, Chu Y, Gao C, Wang H, Chen G, Luo C, Wang T, Chen X, Zhang M, Tao L. Exogenous Hydrogen Sulfide Offers Neuroprotection on Intracerebral Hemorrhage Injury Through Modulating Endogenous H 2S Metabolism in Mice. Front Cell Neurosci 2019; 13:349. [PMID: 31440142 PMCID: PMC6693577 DOI: 10.3389/fncel.2019.00349] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 07/15/2019] [Indexed: 11/21/2022] Open
Abstract
Hydrogen sulfide (H2S), an important endogenous signaling molecule, has a significant neuroprotective role in the central nervous system. In this study, we examined the protective effects of exogenous H2S against intracerebral hemorrhage (ICH), as well as its underlying mechanisms. We investigated the effects of exogenous H2S on ICH using Western blotting, injury volume, measurement of brain edema, propidium iodide (PI) staining, and behavior assessment, respectively. We found that endogenous H2S production was downregulated in the brain after ICH, which is caused by the decrease in cystathionine β-synthase (CBS) as the predominant cerebral H2S-generating enzyme in the brain. Treatment with sodium hydrosulfide (NaHS; an H2S producer) could restore the H2S production and the expression of CBS. NaHS could also attenuate brain edema, injury volume, and neurological deficits in the Morris water maze test after ICH. Western blotting results indicated that H2S pretreatment reversed the increase in caspase 3 cleavage and the decrease in Bcl-2, suppressed the activation of autophagy marker (LC3II and Beclin-1), and maintained the p62 level in injured striatum post-ICH. However, H2S could not restore brain CBS expression and H2S content, reduce brain edema, and improve motor performance and memory function after ICH through modulating autophagy and apoptosis when pretreated with the CBS inhibitor aminooxyacetic acid (AOAA). We also found that AOAA reduced the endogenous H2S production through inhibiting the enzyme activity of CBS rather than modulating the expression of CBS protein level. These present results indicate that H2S may possess potential therapeutic value in the treatment of brain injury after ICH, and the protective effect of exogenous H2S against ICH may be not a direct action but an indirect effect through inducing endogenous H2S metabolism responses.
Collapse
Affiliation(s)
- Haiyan Shan
- Institute of Forensic Sciences, Soochow University, Suzhou, China.,Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jianping Qiu
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Pan Chang
- Central Laboratory, The Second Affiliated Hospital of Xi'an Medical College, Xi'an, China
| | - Yang Chu
- Institute of Forensic Sciences, Soochow University, Suzhou, China
| | - Cheng Gao
- Institute of Forensic Sciences, Soochow University, Suzhou, China
| | - Haocheng Wang
- Institute of Forensic Sciences, Soochow University, Suzhou, China
| | - Guang Chen
- Institute of Forensic Sciences, Soochow University, Suzhou, China
| | - Chengliang Luo
- Institute of Forensic Sciences, Soochow University, Suzhou, China
| | - Tao Wang
- Institute of Forensic Sciences, Soochow University, Suzhou, China
| | - Xiping Chen
- Institute of Forensic Sciences, Soochow University, Suzhou, China
| | - Mingyang Zhang
- Institute of Forensic Sciences, Soochow University, Suzhou, China.,School of Pharmacy, Soochow University, Suzhou, China
| | - Luyang Tao
- Institute of Forensic Sciences, Soochow University, Suzhou, China
| |
Collapse
|
41
|
Qi R, Zhang X, Xie Y, Jiang S, Liu Y, Liu X, Xie W, Jia X, Bade R, Shi R, Li S, Ren C, Gong K, Zhang C, Shao G. 5-Aza-2'-deoxycytidine increases hypoxia tolerance-dependent autophagy in mouse neuronal cells by initiating the TSC1/mTOR pathway. Biomed Pharmacother 2019; 118:109219. [PMID: 31325707 DOI: 10.1016/j.biopha.2019.109219] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Our previous study found that 5-Aza-2'-deoxycytidine (5-Aza-CdR) can repress the expression and activity of protein serine/threonine phosphatase-1γ (PP1γ) in mouse hippocampus. It is well known that PP1γ regulates cell metabolism, which is related to hypoxia/ischaemia tolerance. It has been reported that it can also induce autophagy in cancer cells. Autophagy is important for maintaining cellular homeostasis associated with metabolism. In this study, we examined whether 5-Aza-CdR increases hypoxia tolerance-dependent autophagy by initiating the TSC1/mTOR/autophagy signalling pathway in neuronal cells. METHODS 5-Aza-CdR was either administered to mice via intracerebroventricular injection (i.c.v) or added to cultured hippocampal-derived neuronal cell line (HT22 cell) in the medium for cell culture. The hypoxia tolerance of mice was measured by hypoxia tolerance time and Perl's iron stain. The mRNA and protein expression levels of tuberous sclerosis complex 1 (TSC1), mammalian target of rapamycin (mTOR) and autophagy marker light chain 3 (LC3) were measured by real-time PCR and western blot. The p-mTOR and p-p70S6k proteins were used as markers for mTOR activity. In addition, the role of autophagy was determined by correlating its intensity with hypoxia tolerance in a time-dependent manner. At the same time, the involvement of the TSC1/mTOR pathway in autophagy was also examined through transfection with TSC1 (hamartin) plasmid. RESULTS 5-Aza-CdR was revealed to increase hypoxia tolerance and induce autophagy, accompanied by an increase in mRNA and protein expression levels of TSC1, reduction in p-mTOR (Ser2448) and p-p70S6k (Thr389) protein levels, and an increase in the ratio of LC3-II/LC3-I in both mouse hippocampus and hippocampal-derived neuronal cell line (HT22). The fluorescence intensity of hamartin was enhanced in the hippocampus of mice exposed to 5-Aza-CdR. Moreover, HT22 cells that over-expressed TSC1 showed more autophagy. CONCLUSIONS 5-Aza-CdR can increase hypoxia tolerance by inducing autophagy by initiating the TSC1/mTOR pathway.
Collapse
Affiliation(s)
- Ruifang Qi
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, School of Basic Medical Science, Capital Medical University, Beijing, China; Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaolu Zhang
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yabin Xie
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shuyuan Jiang
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - You Liu
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaolei Liu
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Xie
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoe Jia
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Rengui Bade
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ruili Shi
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Sijie Li
- Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kerui Gong
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, USA
| | - Chunyang Zhang
- Department of neurosurgery, the First Affiliated Hospital of Baotou Medical College, Inner Mongolia, China
| | - Guo Shao
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, School of Basic Medical Science, Capital Medical University, Beijing, China; Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, China; Beijing key laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
42
|
Walsh KB, Zhang X, Zhu X, Wohleb E, Woo D, Lu L, Adeoye O. Intracerebral Hemorrhage Induces Inflammatory Gene Expression in Peripheral Blood: Global Transcriptional Profiling in Intracerebral Hemorrhage Patients. DNA Cell Biol 2019; 38:660-669. [PMID: 31120332 PMCID: PMC6909779 DOI: 10.1089/dna.2018.4550] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/03/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023] Open
Abstract
To perform global transcriptome profiling using RNA-seq in the peripheral blood of intracerebral hemorrhage (ICH) patients. In 11 patients with ICH, peripheral blood was collected within 24 h of symptom onset or last known well, and a second blood draw occurred 72 h (±6) after the first. RNA-seq identified differentially expressed genes (DEGs) between the first and second samples. Biological pathway enrichment analysis was performed with Ingenuity® Pathway Analysis (IPA). A total of 16,640 genes were identified and 218 were significant DEGs after ICH (false discovery rate <0.1). IPA identified 97 disease and functional categories that were significantly upregulated (z-score >2) post-ICH; 46 categories were specifically related to immune cell activation, 22 to general cellular activation processes, and 4 to other inflammation-related responses. In the canonical pathway and network analysis, inflammatory mediators of particular importance included interleukin-8, NF-κB, ERK1/2, and members of the integrin class. ICH induced peripheral blood gene expression at 72 to 96 h compared with 0 to 24 h from symptom onset. DEGs that were highly expressed included those related to inflammation and activation of the immune response. Further research is needed to determine whether these changes affect outcomes and may represent new therapeutic targets.
Collapse
Affiliation(s)
- Kyle B. Walsh
- University of Cincinnati Gardner Neuroscience Institute, Cincinnati, Ohio
- Department of Emergency Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Xiang Zhang
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio
| | - Xiaoting Zhu
- Division of Biomedical Informatics, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| | - Eric Wohleb
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio
- University of Cincinnati Neurobiology Research Center, Cincinnati, Ohio
| | - Daniel Woo
- University of Cincinnati Gardner Neuroscience Institute, Cincinnati, Ohio
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Long Lu
- Division of Biomedical Informatics, Cincinnati Children's Research Foundation, Cincinnati, Ohio
| | - Opeolu Adeoye
- University of Cincinnati Gardner Neuroscience Institute, Cincinnati, Ohio
- Department of Emergency Medicine, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
43
|
Different intensity of autophagy regulate interleukin-33 to control the uncontrolled inflammation of acute lung injury. Inflamm Res 2019; 68:665-675. [PMID: 31147742 DOI: 10.1007/s00011-019-01250-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES Cytokines participate in the progression of acute respiratory distress syndrome (ARDS), and uncontrolled inflammation is a central issue of acute lung injury (ALI). Interleukin (IL)-33 is a nuclear protein that has been reported to have a proinflammatory role in ARDS. Studies have shown that excessive autophagy may lead to the increased mortality of patients with ARDS, while several investigations indicated that IL-33 and autophagy interact with one another. The present study sought to clarify the relation between autophagy and IL-33's proinflammatory role in ARDS. METHODS We built a lipopolysaccharide (LPS)-induced lung injury mouse model. To study the relationship between IL-33 and autophagy, mice were pretreated with rapamycin (RAPA; a promoter of autophagy) and 3-methyladenine (3-MA; an inhibitor of autophagy) prior to LPS administration. The expression of IL-33 in serum and bronchoalveolar lavage fluid (BALF) was measured. Immunohistochemistry of IL-33 in lung tissue was examined. Th1,Th2 cytokines/chemokine levels in serum and BALF were tested. Further, the severity of lung injury was evaluated. And the nuclear factor-kappa B (NF-κB)'s nuclear translocation in lung tissue was detected. RESULTS In comparison with the control group, the levels of IL-33 in serum and BALF were increased after LPS injection. Th1 cytokines/chemokine levels were significantly increased in serum and BALF, while Th2 cytokine levels changed only a little. The levels of IL-33 in serum and BALF of the RAPA group was significantly increased after LPS was injected as compared with the LPS group; additionally, the levels of IL-33 in serum and BALF of the 3-MA group was significantly reduced after LPS was injected as compared with the LPS group, and that lung injury was ameliorated after 3-MA pretreatment. Th1 cytokines and chemokines in both serum and BALF were also decreased in the 3-MA group. Furthermore, we found that the nuclear translocation of NF-κB increased after LPS administration, and NF-κB's nuclear translocation was significantly increased in comparison with the LPS group after RAPA pretreatment. In contrast, NF-κB's nuclear translocation decreased after 3-MA pretreatment as compared with the LPS group. CONCLUSIONS These findings showed that autophagy might regulate IL-33 by activating or inhibiting NF-κB to control the uncontrolled inflammation of acute lung injury.
Collapse
|
44
|
Durocher M, Ander BP, Jickling G, Hamade F, Hull H, Knepp B, Liu DZ, Zhan X, Tran A, Cheng X, Ng K, Yee A, Sharp FR, Stamova B. Inflammatory, regulatory, and autophagy co-expression modules and hub genes underlie the peripheral immune response to human intracerebral hemorrhage. J Neuroinflammation 2019; 16:56. [PMID: 30836997 PMCID: PMC6399982 DOI: 10.1186/s12974-019-1433-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 02/12/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) has a high morbidity and mortality. The peripheral immune system and cross-talk between peripheral blood and brain have been implicated in the ICH immune response. Thus, we delineated the gene networks associated with human ICH in the peripheral blood transcriptome. We also compared the differentially expressed genes in blood following ICH to a prior human study of perihematomal brain tissue. METHODS We performed peripheral blood whole-transcriptome analysis of ICH and matched vascular risk factor control subjects (n = 66). Gene co-expression network analysis identified groups of co-expressed genes (modules) associated with ICH and their most interconnected genes (hubs). Mixed-effects regression identified differentially expressed genes in ICH compared to controls. RESULTS Of seven ICH-associated modules, six were enriched with cell-specific genes: one neutrophil module, one neutrophil plus monocyte module, one T cell module, one Natural Killer cell module, and two erythroblast modules. The neutrophil/monocyte modules were enriched in inflammatory/immune pathways; the T cell module in T cell receptor signaling genes; and the Natural Killer cell module in genes regulating alternative splicing, epigenetic, and post-translational modifications. One erythroblast module was enriched in autophagy pathways implicated in experimental ICH, and NRF2 signaling implicated in hematoma clearance. Many hub genes or module members, such as IARS, mTOR, S1PR1, LCK, FYN, SKAP1, ITK, AMBRA1, NLRC4, IL6R, IL17RA, GAB2, MXD1, PIK3CD, NUMB, MAPK14, DDX24, EVL, TDP1, ATG3, WDFY3, GSK3B, STAT3, STX3, CSF3R, PIP4K2A, ANXA3, DGAT2, LRP10, FLOT2, ANK1, CR1, SLC4A1, and DYSF, have been implicated in neuroinflammation, cell death, transcriptional regulation, and some as experimental ICH therapeutic targets. Gene-level analysis revealed 1225 genes (FDR p < 0.05, fold-change > |1.2|) have altered expression in ICH in peripheral blood. There was significant overlap of the 1225 genes with dysregulated genes in human perihematomal brain tissue (p = 7 × 10-3). Overlapping genes were enriched for neutrophil-specific genes (p = 6.4 × 10-08) involved in interleukin, neuroinflammation, apoptosis, and PPAR signaling. CONCLUSIONS This study delineates key processes underlying ICH pathophysiology, complements experimental ICH findings, and the hub genes significantly expand the list of novel ICH therapeutic targets. The overlap between blood and brain gene responses underscores the importance of examining blood-brain interactions in human ICH.
Collapse
Affiliation(s)
- Marc Durocher
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Bradley P. Ander
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Glen Jickling
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Farah Hamade
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Heather Hull
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Bodie Knepp
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Da Zhi Liu
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Xinhua Zhan
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Anh Tran
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Xiyuan Cheng
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Kwan Ng
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Alan Yee
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Frank R. Sharp
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
| | - Boryana Stamova
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA 95817 USA
- MIND Institute Biosciences Building, 2805 50th Street, Sacramento, CA 95817 USA
| |
Collapse
|
45
|
Bimodal Distribution of Nuclear Factor-κB Activation and Expression of Subunits in Experimental Models of Intracerebral Hemorrhage In Vivo. J Stroke Cerebrovasc Dis 2019; 28:821-829. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.11.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/12/2018] [Accepted: 11/29/2018] [Indexed: 02/08/2023] Open
|
46
|
Zhang X, Fu C, Chen B, Xu Z, Zeng Z, He L, Lu Y, Chen Z, Liu X. Autophagy Induced by Oxygen-Glucose Deprivation Mediates the Injury to the Neurovascular Unit. Med Sci Monit 2019; 25:1373-1382. [PMID: 30787267 PMCID: PMC6394142 DOI: 10.12659/msm.915123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Autophagy is characterized by the degradation of cellular components in autophagosomes. It plays a significant role in cerebral ischemic injury and has a complex functional connection with apoptosis. The neurovascular unit (NVU) is a structural and functional unit of the nervous system presented as a therapeutic target of stroke. This study aimed to investigate the effect of autophagy induced by ischemic damage on NVUs. MATERIAL AND METHODS SH-SY5Y cells, C6 cells, and rat brain microvascular endothelial cells were cultured with oxygen-glucose deprivation (OGD) exposure for different time durations, and 3-methyladenine (3-MA) was added as an autophagy inhibitor. In all 3 cell lines, lactate dehydrogenase (LDH) release was measured. Furthermore, apoptosis was detected using Annexin V-fluorescein isothiocyanate/propidium iodide labeling and immunofluorescence staining. Autophagosomes were observed through AO/MDC (acridine orange/monodansycadaverine) double staining. LC3-II expression levels were evaluated by western blot analysis. RESULTS In the OGD groups of 3 cell lines, LDH leakage, and apoptotic rates were obviously increased. Remarkable increase in LC3-II expression was found in the OGD groups of SH-SY5Y cells and C6 cells. However, 3-MA decreased the LC3-II expression to varying degrees. CONCLUSIONS OGD could induce the over-activation of autophagy and augment the apoptotic activity in neurons and glial cells of NVUs.
Collapse
Affiliation(s)
- Xinyang Zhang
- Beijing University of Chinese Medicine, Beijing, China (mainland)
| | - Chen Fu
- Central Laboratory, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China (mainland)
| | - Baoxin Chen
- Department of Neurology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China (mainland)
| | - Zhenmin Xu
- Beijing University of Chinese Medicine, Beijing, China (mainland)
| | - Zixiu Zeng
- Beijing University of Chinese Medicine, Beijing, China (mainland)
| | - Lijuan He
- Department of Neurology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China (mainland)
| | - Yan Lu
- Department of Neurology, Xiyuan Hospital, China Academy of Chinese Medical Science, Beijing, China (mainland)
| | - Zhigang Chen
- Department of Neurology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China (mainland)
| | - Xuemei Liu
- Central Laboratory, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China (mainland)
| |
Collapse
|
47
|
Jia J, Zhang M, Li Q, Zhou Q, Jiang Y. Long noncoding ribonucleic acid NKILA induces the endoplasmic reticulum stress/autophagy pathway and inhibits the nuclear factor‐k‐gene binding pathway in rats after intracerebral hemorrhage. J Cell Physiol 2018; 233:8839-8849. [PMID: 29893407 DOI: 10.1002/jcp.26798] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/30/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Jiaoying Jia
- Department of Neurosurgery The Second Xiangya Hospital of Central South University Changsha China
| | - Mingming Zhang
- Department of Neurosurgery The Second Xiangya Hospital of Central South University Changsha China
| | - Qi Li
- Department of Neurosurgery The Second Xiangya Hospital of Central South University Changsha China
| | - Qian Zhou
- Department of Neurosurgery The Second Xiangya Hospital of Central South University Changsha China
| | - Yugang Jiang
- Department of Neurosurgery The Second Xiangya Hospital of Central South University Changsha China
| |
Collapse
|
48
|
Lopez JR, Uryash A, Kolster J, Estève E, Zhang R, Adams JA. Enhancing Endogenous Nitric Oxide by Whole Body Periodic Acceleration Elicits Neuroprotective Effects in Dystrophic Neurons. Mol Neurobiol 2018; 55:8680-8694. [DOI: 10.1007/s12035-018-1018-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 03/16/2018] [Indexed: 12/25/2022]
|
49
|
Wu Y, Wang L, Hu K, Yu C, Zhu Y, Zhang S, Shao A. Mechanisms and Therapeutic Targets of Depression After Intracerebral Hemorrhage. Front Psychiatry 2018; 9:682. [PMID: 30618863 PMCID: PMC6304443 DOI: 10.3389/fpsyt.2018.00682] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/23/2018] [Indexed: 12/17/2022] Open
Abstract
The relationship between depression and intracerebral hemorrhage (ICH) is complicated. One of the most common neuropsychiatric comorbidities of hemorrhagic stroke is Post-ICH depression. Depression, as a neuropsychiatric symptom, also negatively impacts the outcome of ICH by enhancing morbidity, disability, and mortality. However, the ICH outcome can be improved by antidepressants such as the frequently-used selective serotonin reuptake inhibitors. This review therefore presents the mechanisms of post-ICH depression, we grouped the mechanisms according to inflammation, oxidative stress (OS), apoptosis and autophagy, and explained them through their several associated signaling pathways. Inflammation is mainly related to Toll-like receptors (TLRs), the NF-kB mediated signal pathway, the PPAR-γ-dependent pathway, as well as other signaling pathways. OS is associated to nuclear factor erythroid-2 related factor 2 (Nrf2), the PI3K/Akt pathway and the MAPK/P38 pathway. Moreover, autophagy is associated with the mTOR signaling cascade and the NF-kB mediated signal pathway, while apoptosis is correlated with the death receptor-mediated apoptosis pathway, mitochondrial apoptosis pathway, caspase-independent pathways and others. Furthermore, we found that neuroinflammation, oxidative stress, autophagy, and apoptosis experience interactions with one another. Additionally, it may provide several potential therapeutic targets for patients that might suffer from depression after ICH.
Collapse
Affiliation(s)
- Yinan Wu
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liangliang Wang
- Interdisciplinary Institute of Neuroscience and Technology, Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, China
| | - Kaimin Hu
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chengcheng Yu
- Department of Orthopedics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanhan Zhu
- Department of Neurosurgery, Rongjun Hospital, Jiaxing, China
| | - Suzhan Zhang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
50
|
Zhao H, Chen Y, Feng H. P2X7 Receptor-Associated Programmed Cell Death in the Pathophysiology of Hemorrhagic Stroke. Curr Neuropharmacol 2018; 16:1282-1295. [PMID: 29766811 PMCID: PMC6251042 DOI: 10.2174/1570159x16666180516094500] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/17/2017] [Accepted: 02/28/2018] [Indexed: 12/20/2022] Open
Abstract
Hemorrhagic stroke is a life-threatening disease characterized by a sudden rupture of cerebral blood vessels, and cell death is widely believed to occur after exposure to blood metabolites or subsequently damaged cells. Recently, programmed cell death, such as apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis, has been demonstrated to play crucial roles in the pathophysiology of stroke. However, the detailed mechanisms of these novel kinds of cell death are still unclear. The P2X7 receptor, previously known for its cytotoxic activity, is an ATP-gated, nonselective cation channel that belongs to the family of ionotropic P2X receptors. Evolving evidence indicates that the P2X7 receptor plays a pivotal role in central nervous system pathology; genetic deletion and pharmacological blockade of the P2X7 receptor provide neuroprotection in various neurological disorders, including intracerebral hemorrhage and subarachnoid hemorrhage. The P2X7 receptor may regulate programmed cell death via (I) exocytosis of secretory lysosomes, (II) exocytosis of autophagosomes or autophagolysosomes during formation of the initial autophagic isolation membrane or omegasome, and (III) direct release of cytosolic IL-1β secondary to regulated cell death by pyroptosis or necroptosis. In this review, we present an overview of P2X7 receptor- associated programmed cell death for further understanding of hemorrhagic stroke pathophysiology, as well as potential therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Hengli Zhao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| |
Collapse
|