1
|
Wang Y, Liu C, Ren Y, Song J, Fan K, Gao L, Ji X, Chen X, Zhao H. Nanomaterial-Based Strategies for Attenuating T-Cell-Mediated Immunodepression in Stroke Patients: Advancing Research Perspectives. Int J Nanomedicine 2024; 19:5793-5812. [PMID: 38882535 PMCID: PMC11180442 DOI: 10.2147/ijn.s456632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 05/14/2024] [Indexed: 06/18/2024] Open
Abstract
This review article discusses the potential of nanomaterials in targeted therapy and immunomodulation for stroke-induced immunosuppression. Although nanomaterials have been extensively studied in various biomedical applications, their specific use in studying and addressing immunosuppression after stroke remains limited. Stroke-induced neuroinflammation is characterized by T-cell-mediated immunodepression, which leads to increased morbidity and mortality. Key observations related to immunodepression after stroke, including lymphopenia, T-cell dysfunction, regulatory T-cell imbalance, and cytokine dysregulation, are discussed. Nanomaterials, such as liposomes, micelles, polymeric nanoparticles, and dendrimers, offer advantages in the precise delivery of drugs to T cells, enabling enhanced targeting and controlled release of immunomodulatory agents. These nanomaterials have the potential to modulate T-cell function, promote neuroregeneration, and restore immune responses, providing new avenues for stroke treatment. However, challenges related to biocompatibility, stability, scalability, and clinical translation need to be addressed. Future research efforts should focus on comprehensive studies to validate the efficacy and safety of nanomaterial-based interventions targeting T cells in stroke-induced immunosuppression. Collaborative interdisciplinary approaches are necessary to advance the field and translate these innovative strategies into clinical practice, ultimately improving stroke outcomes and patient care.
Collapse
Grants
- This work was supported by the National Natural Science Foundation of China (Grant number 82001248), National University of Singapore (NUHSRO/2020/133/Startup/08, NUHSRO/2023/008/NUSMed/TCE/LOA, NUHSRO/2021/034/TRP/09/Nanomedicine, NUHSRO/2021/044/Kickstart/09/LOA, 23-0173-A0001), National Medical Research Council (MOH-001388-00, CG21APR1005, OFIRG23jul-0047), Singapore Ministry of Education (MOE-000387-00), and National Research Foundation (NRF-000352-00)
Collapse
Affiliation(s)
- Yan Wang
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Cuiying Liu
- School of Nursing, Capital Medical University, Beijing, People’s Republic of China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Yanhong Ren
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Jibin Song
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysics Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Heng Zhao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
2
|
DeLong JH, Ohashi SN, O'Connor KC, Sansing LH. Inflammatory Responses After Ischemic Stroke. Semin Immunopathol 2022; 44:625-648. [PMID: 35767089 DOI: 10.1007/s00281-022-00943-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/20/2022] [Indexed: 12/25/2022]
Abstract
Ischemic stroke generates an immune response that contributes to neuronal loss as well as tissue repair. This is a complex process involving a range of cell types and effector molecules and impacts tissues outside of the CNS. Recent reviews address specific aspects of this response, but several years have passed and important advances have been made since a high-level review has summarized the overall state of the field. The present review examines the initiation of the inflammatory response after ischemic stroke, the complex impacts of leukocytes on patient outcome, and the potential of basic science discoveries to impact the development of therapeutics. The information summarized here is derived from broad PubMed searches and aims to reflect recent research advances in an unbiased manner. We highlight valuable recent discoveries and identify gaps in knowledge that have the potential to advance our understanding of this disease and therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Jonathan Howard DeLong
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Sarah Naomi Ohashi
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin Charles O'Connor
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Lauren Hachmann Sansing
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
Wang X, Wang Q, Wang K, Ni Q, Li H, Su Z, Xu Y. Is Immune Suppression Involved in the Ischemic Stroke? A Study Based on Computational Biology. Front Aging Neurosci 2022; 14:830494. [PMID: 35250546 PMCID: PMC8896355 DOI: 10.3389/fnagi.2022.830494] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/20/2022] [Indexed: 01/01/2023] Open
Abstract
Objective To identify the genetic mechanisms of immunosuppression-related genes implicated in ischemic stroke. Background A better understanding of immune-related genes (IGs) involved in the pathophysiology of ischemic stroke may help identify drug targets beneficial for immunomodulatory approaches and reducing stroke-induced immunosuppression complications. Methods Two datasets related to ischemic stroke were downloaded from the GEO database. Immunosuppression-associated genes were obtained from three databases (i.e., DisGeNET, HisgAtlas, and Drugbank). The CIBERSORT algorithm was used to calculate the mean proportions of 22 immune-infiltrating cells in the stroke samples. Differential gene expression analysis was performed to identify the differentially expressed genes (DEGs) involved in stroke. Immunosuppression-related crosstalk genes were identified as the overlapping genes between ischemic stroke-DEGs and IGs. Feature selection was performed using the Boruta algorithm and a classifier model was constructed to evaluate the prediction accuracy of the obtained immunosuppression-related crosstalk genes. Functional enrichment analysis, gene-transcriptional factor and gene-drug interaction networks were constructed. Results Twenty two immune cell subsets were identified in stroke, where resting CD4 T memory cells were significantly downregulated while M0 macrophages were significantly upregulated. By overlapping the 54 crosstalk genes obtained by feature selection with ischemic stroke-related genes obtained from the DisGenet database, 17 potentially most valuable immunosuppression-related crosstalk genes were obtained, ARG1, CD36, FCN1, GRN, IL7R, JAK2, MAFB, MMP9, PTEN, STAT3, STAT5A, THBS1, TLR2, TLR4, TLR7, TNFSF10, and VASP. Regulatory transcriptional factors targeting key immunosuppression-related crosstalk genes in stroke included STAT3, SPI1, CEPBD, SP1, TP53, NFIL3, STAT1, HIF1A, and JUN. In addition, signaling pathways enriched by the crosstalk genes, including PD-L1 expression and PD-1 checkpoint pathway, NF-kappa B signaling, IL-17 signaling, TNF signaling, and NOD-like receptor signaling, were also identified. Conclusion Putative crosstalk genes that link immunosuppression and ischemic stroke were identified using bioinformatics analysis and machine learning approaches. These may be regarded as potential therapeutic targets for ischemic stroke.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qian Wang
- Postdoctoral Workstation, Taian City Central Hospital, Taian, China
| | - Kun Wang
- Postdoctoral Workstation, Taian City Central Hospital, Taian, China
| | - Qingbin Ni
- Postdoctoral Workstation, Taian City Central Hospital, Taian, China
| | - Hu Li
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Zhiqiang Su
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
- *Correspondence: Yuzhen Xu,
| |
Collapse
|
4
|
Liu H, Zhan F, Wang Y. Evaluation of monocyte-to-high-density lipoprotein cholesterol ratio and monocyte-to-lymphocyte ratio in ischemic stroke. J Int Med Res 2021; 48:300060520933806. [PMID: 32660292 PMCID: PMC7361497 DOI: 10.1177/0300060520933806] [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] [Indexed: 01/01/2023] Open
Abstract
Objectives We aimed to evaluate the diagnostic value of the combination of the monocyte-to-high-density lipoprotein cholesterol ratio (MHR) with the monocyte-to-lymphocyte ratio (MLR) in ischemic stroke patients. Methods There were 253 patients who were diagnosed with ischemic stroke and 211 healthy subjects enrolled into this retrospective study. Result MHR and MLR were significantly higher in ischemic stroke patients compared with controls. MHR and MLR remained as independent variables for the presence of ischemic stroke. In receiver operating characteristic analyses, the optimal cut-off values for MHR and MLR were 0.28 and 0.19, respectively. The area under the curve for MHR was 0.777 (sensitivity, 66.01%; specificity, 77.25%), and that for MLR was 0.742 (sensitivity, 70.36%; specificity, 67.77%) in ischemic stroke patients. Moreover, the combination MHR and MLR increased the sensitivity compared with MHR or MLR alone. Conclusion The present study shows that a high MHR and MLR are each predictive for the risk of ischemic stroke, and together, they exhibit a better diagnostic value compared with each ratio alone.
Collapse
Affiliation(s)
- Huiling Liu
- Department of Clinical Laboratory, Changzhou Tumor Hospital Affiliated with Soochow University, Jiangsu, China
| | - Feng Zhan
- Department of Clinical Laboratory, Changzhou Tumor Hospital Affiliated with Soochow University, Jiangsu, China
| | - Yazhou Wang
- Department of Clinical Laboratory, Changzhou Tumor Hospital Affiliated with Soochow University, Jiangsu, China
| |
Collapse
|
5
|
Lei TY, Ye YZ, Zhu XQ, Smerin D, Gu LJ, Xiong XX, Zhang HF, Jian ZH. The immune response of T cells and therapeutic targets related to regulating the levels of T helper cells after ischaemic stroke. J Neuroinflammation 2021; 18:25. [PMID: 33461586 PMCID: PMC7814595 DOI: 10.1186/s12974-020-02057-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/09/2020] [Indexed: 12/21/2022] Open
Abstract
Through considerable effort in research and clinical studies, the immune system has been identified as a participant in the onset and progression of brain injury after ischaemic stroke. Due to the involvement of all types of immune cells, the roles of the immune system in stroke pathology and associated effects are complicated. Past research concentrated on the functions of monocytes and neutrophils in the pathogenesis of ischaemic stroke and tried to demonstrate the mechanisms of tissue injury and protection involving these immune cells. Within the past several years, an increasing number of studies have elucidated the vital functions of T cells in the innate and adaptive immune responses in both the acute and chronic phases of ischaemic stroke. Recently, the phenotypes of T cells with proinflammatory or anti-inflammatory function have been demonstrated in detail. T cells with distinctive phenotypes can also influence cerebral inflammation through various pathways, such as regulating the immune response, interacting with brain-resident immune cells and modulating neurogenesis and angiogenesis during different phases following stroke. In view of the limited treatment options available following stroke other than tissue plasminogen activator therapy, understanding the function of immune responses, especially T cell responses, in the post-stroke recovery period can provide a new therapeutic direction. Here, we discuss the different functions and temporal evolution of T cells with different phenotypes during the acute and chronic phases of ischaemic stroke. We suggest that modulating the balance between the proinflammatory and anti-inflammatory functions of T cells with distinct phenotypes may become a potential therapeutic approach that reduces the mortality and improves the functional outcomes and prognosis of patients suffering from ischaemic stroke.
Collapse
Affiliation(s)
- Tian-Yu Lei
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Ying-Ze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Xi-Qun Zhu
- Department of Head and Neck and Neurosurgery, Hubei Cancer Hospital, Wuhan, 430079, Hubei Province, People's Republic of China
| | - Daniel Smerin
- University of Central Florida College of Medicine, Orlando, FL, 32827, USA
| | - Li-Juan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Xiao-Xing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Zhi-Hong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
6
|
Kong Y, Li S, Cheng X, Ren H, Zhang B, Ma H, Li M, Zhang XA. Brain Ischemia Significantly Alters microRNA Expression in Human Peripheral Blood Natural Killer Cells. Front Immunol 2020; 11:759. [PMID: 32477329 PMCID: PMC7240012 DOI: 10.3389/fimmu.2020.00759] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 04/03/2020] [Indexed: 11/13/2022] Open
Abstract
Brain ischemia induces systemic immunosuppression and increases a host's susceptibility to infection. MicroRNAs (miRNAs) are molecular switches in immune cells, but the alterations of miRNAs in human immune cells in response to brain ischemia and their impact on immune defense remain elusive. Natural killer (NK) cells are critical for early host defenses against pathogens. In this study, we identified reduced counts, cytokine production, and cytotoxicity in human peripheral blood NK cells obtained from patients with acute ischemic stroke. The extent of NK cell loss of number and activity was associated with infarct volume. MicroRNA sequencing analysis revealed that brain ischemia significantly altered miRNA expression profiles in circulating NK cells, in which miRNA-451a and miRNA-122-5p were dramatically upregulated. Importantly, inhibition of miR-451a or miR-122-5p augmented the expression of activation-associated receptors in NK cells. These results provide the first evidence that brain ischemia alters miRNA signatures in human NK cells.
Collapse
Affiliation(s)
- Ying Kong
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Shiyao Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Xiaojing Cheng
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Honglei Ren
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.,Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Bohao Zhang
- Department of Imaging, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongshan Ma
- Center for Neurological Diseases, The Third People's Hospital of Datong, Datong, China
| | - Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-neurotrauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Xiao-An Zhang
- Department of Imaging, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Wang Y, Luo Y, Yao Y, Ji Y, Feng L, Du F, Zheng X, Tao T, Zhai X, Li Y, Han P, Xu B, Zhao H. Silencing the lncRNA Maclpil in pro-inflammatory macrophages attenuates acute experimental ischemic stroke via LCP1 in mice. J Cereb Blood Flow Metab 2020; 40:747-759. [PMID: 30895879 PMCID: PMC7168792 DOI: 10.1177/0271678x19836118] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Long noncoding RNAs (lncRNA) expression profiles change in the ischemic brain after stroke, but their roles in specific cell types after stroke have not been studied. We tested the hypothesis that lncRNA modulates brain injury by altering macrophage functions. Using RNA deep sequencing, we identified 73 lncRNAs that were differentially expressed in monocyte-derived macrophages (MoDMs) and microglia-derived macrophages (MiDMs) isolated in the ischemic brain three days after stroke. Among these, the lncRNA, GM15628, is highly expressed in pro-inflammatory MoDMs but not in MiDMs, and are functionally related to its neighbor gene, lymphocyte cytosolic protein 1 (LCP1), which plays a role in maintaining cell shape and cell migration. We termed this lncRNA as Macrophage contained LCP1 related pro-inflammatory lncRNA, Maclpil. Using cultured macrophages polarized by LPS, M(LPS), we found that downregulation of Maclpil in M(LPS) decreased pro-inflammatory gene expression while promoting anti-inflammatory gene expression. Maclpil inhibition also reduced the migration and phagocytosis ability of MoDMs by inhibiting LCP1. Furthermore, adoptive transfer of Maclpil silenced M(LPS), reduced ischemic brain infarction, improved behavioral performance and attenuated penetration of MoDMs in the ischemic hemisphere. We conclude that by blocking macrophage, Maclpil protects against acute ischemic stroke by inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Yan Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ying Luo
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yang Yao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuhua Ji
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Liangshu Feng
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Fang Du
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoya Zheng
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tao Tao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xuan Zhai
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yaning Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Pei Han
- Department of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
8
|
Ye Y, Zeng Z, Jin T, Zhang H, Xiong X, Gu L. The Role of High Mobility Group Box 1 in Ischemic Stroke. Front Cell Neurosci 2019; 13:127. [PMID: 31001089 PMCID: PMC6454008 DOI: 10.3389/fncel.2019.00127] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 03/14/2019] [Indexed: 12/11/2022] Open
Abstract
High-mobility group box 1 protein (HMGB1) is a novel, cytokine-like, and ubiquitous, highly conserved, nuclear protein that can be actively secreted by microglia or passively released by necrotic neurons. Ischemic stroke is a leading cause of death and disability worldwide, and the outcome is dependent on the amount of hypoxia-related neuronal death in the cerebral ischemic region. Acting as an endogenous danger-associated molecular pattern (DAMP) protein, HMGB1 mediates cerebral inflammation and brain injury and participates in the pathogenesis of ischemic stroke. It is thought that HMGB1 signals via its presumed receptors, such as toll-like receptors (TLRs), matrix metalloproteinase (MMP) enzymes, and receptor for advanced glycation end products (RAGEs) during ischemic stroke. In addition, the release of HMGB1 from the brain into the bloodstream influences peripheral immune cells. However, the role of HMGB1 in ischemic stroke may be more complex than this and has not yet been clarified. Here, we summarize and review the research into HMGB1 in ischemic stroke.
Collapse
Affiliation(s)
- Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tong Jin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongfei Zhang
- Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Xiaoxing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Wang Z, He D, Zeng YY, Zhu L, Yang C, Lu YJ, Huang JQ, Cheng XY, Huang XH, Tan XJ. The spleen may be an important target of stem cell therapy for stroke. J Neuroinflammation 2019; 16:20. [PMID: 30700305 PMCID: PMC6352449 DOI: 10.1186/s12974-019-1400-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022] Open
Abstract
Stroke is the most common cerebrovascular disease, the second leading cause of death behind heart disease and is a major cause of long-term disability worldwide. Currently, systemic immunomodulatory therapy based on intravenous cells is attracting attention. The immune response to acute stroke is a major factor in cerebral ischaemia (CI) pathobiology and outcomes. Over the past decade, the significant contribution of the spleen to ischaemic stroke has gained considerable attention in stroke research. The changes in the spleen after stroke are mainly reflected in morphology, immune cells and cytokines, and these changes are closely related to the stroke outcomes. Autonomic nervous system (ANS) activation, release of central nervous system (CNS) antigens and chemokine/chemokine receptor interactions have been documented to be essential for efficient brain-spleen cross-talk after stroke. In various experimental models, human umbilical cord blood cells (hUCBs), haematopoietic stem cells (HSCs), bone marrow stem cells (BMSCs), human amnion epithelial cells (hAECs), neural stem cells (NSCs) and multipotent adult progenitor cells (MAPCs) have been shown to reduce the neurological damage caused by stroke. The different effects of these cell types on the interleukin (IL)-10, interferon (IFN), and cholinergic anti-inflammatory pathways in the spleen after stroke may promote the development of new cell therapy targets and strategies. The spleen will become a potential target of various stem cell therapies for stroke represented by MAPC treatment.
Collapse
Affiliation(s)
- Zhe Wang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China.,Institute of Reproductive and Stem Cell Research, School of Basic Medical Science, Central South University, Changsha, 410000, China
| | - Da He
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Ya-Yue Zeng
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Li Zhu
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Chao Yang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Yong-Juan Lu
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Jie-Qiong Huang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiao-Yan Cheng
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiang-Hong Huang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiao-Jun Tan
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China.
| |
Collapse
|
10
|
Zhang H, Xiong X, Gu L, Xie W, Zhao H. CD4 T cell deficiency attenuates ischemic stroke, inhibits oxidative stress, and enhances Akt/mTOR survival signaling pathways in mice. Chin Neurosurg J 2018; 4. [PMID: 32832192 PMCID: PMC7398241 DOI: 10.1186/s41016-018-0140-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background Inhibition of CD4 T cells reduces stroke-induced infarction by inhibiting neuroinflammation in the ischemic brain in experimental stroke. Nevertheless, little is known about its effects on neuronal survival signaling pathways. In this study, we investigated the effects of CD4 T cell deficits on oxidative stress and on the Akt/mTOR cell signaling pathways after ischemic stroke in mice. Methods MHC II gene knockout C57/BL6 mice, with significantly decreased CD4 T cells, were used. Stroke was induced by 60-min middle cerebral artery (MCA) occlusion. Ischemic brain tissues were harvested for Western blotting. Results The impairment of CD4 T cell production resulted in smaller infarction. The Western blot results showed that iNOS protein levels robustly increased at 5 h and 24 h and then returned toward baseline at 48 h in wild-type mice after stroke, and gene KO inhibited iNOS at 5 h and 24 h. In contrast, the anti-inflammatory marker, arginase I, was found increased after stroke in WT mice, which was further enhanced in the KO mice. In addition, stroke resulted in increased phosphorylated PTEN, Akt, PRAS40, P70S6, and S6 protein levels in WT mice, which were further enhanced in the animals whose CD4 T cells were impaired. Conclusion The impairment of CD4 T cell products prevents ischemic brain injury, inhibits inflammatory signals, and enhances the Akt/mTOR cell survival signaling pathways.
Collapse
Affiliation(s)
- Hongfei Zhang
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Rd., MSLS Bldg., Room P306, Stanford, CA 94305, USA.,Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Rd., MSLS Bldg., Room P306, Stanford, CA 94305, USA.,Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lijuan Gu
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Rd., MSLS Bldg., Room P306, Stanford, CA 94305, USA.,Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Weiying Xie
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Rd., MSLS Bldg., Room P306, Stanford, CA 94305, USA.,Department of Anesthesiology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, 1201 Welch Rd., MSLS Bldg., Room P306, Stanford, CA 94305, USA
| |
Collapse
|
11
|
Ibrahim EH, Kilany M, Ghramh HA, Khan KA, Ul Islam S. Cellular proliferation/cytotoxicity and antimicrobial potentials of green synthesized silver nanoparticles (AgNPs) using Juniperus procera. Saudi J Biol Sci 2018; 26:1689-1694. [PMID: 31762645 PMCID: PMC6864202 DOI: 10.1016/j.sjbs.2018.08.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/09/2018] [Accepted: 08/14/2018] [Indexed: 12/18/2022] Open
Abstract
Juniperus spp. are used as medicinal plants in many countries like Bosnia, Lebanon, and Turkey. In folk medicines, these plants have been used for treating skin and respiratory tract diseases, urinary problems, rheumatism and gall bladder stones. The objectives of this work were to synthesize silver nanoparticles (AgNPs) using a coniferous tree, Juniperus procera leaf extract and testing the synthesized AgNPs for its antimicrobial potentials, hemolytic activity, toxicity and the proliferative effects against normal and activated rat splenic cells. Leaf extract was prepared using acetone and ethanol as solvents. AgNPs were prepared using the acetone extract. AgNPs were validated using UV–Vis spectroscopy and scanning electron microscopy (SEM). Functional groups in the extract were identified using Fourier Transform Infrared (FT-IR) spectroscopy. SEM images of AgNPs showed spherical and cubic shapes with a uniform size distribution with an average size of 30–90 nm. FT-IR spectroscopy showed the presence of many functional groups in the plant extract. AgNPs showed promising antimicrobial activity against tested bacteria and fungus. AgNPs also expressed a stimulating activity towards the rat splenic cells in a dose dependent manner. Acetone as solvent was safer on cells than ethanol. Green synthesized AgNPs using J. procera might be used as a broad-spectrum therapeutic agent against microorganisms and as an immunostimulant agent.
Collapse
Affiliation(s)
- Essam H Ibrahim
- Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia.,Blood Products Quality Control and Research Department, National Organization for Research and Control of Biologicals, Cairo, Egypt
| | - Mona Kilany
- Biology Department, Faculty of Sciences and Arts, King Khalid University, Dhahran Al Janoub, Saudi Arabia.,Department of Microbiology, National Organization for Drug Control and Research (NODCAR), Cairo, Egypt
| | - Hamed A Ghramh
- Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia.,Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia.,Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Khalid Ali Khan
- Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia.,Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Saif Ul Islam
- College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
12
|
Gandolfi M, Smania N, Vella A, Picelli A, Chirumbolo S. Assessed and Emerging Biomarkers in Stroke and Training-Mediated Stroke Recovery: State of the Art. Neural Plast 2017; 2017:1389475. [PMID: 28373915 PMCID: PMC5360976 DOI: 10.1155/2017/1389475] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/11/2017] [Indexed: 12/13/2022] Open
Abstract
Since the increasing update of the biomolecular scientific literature, biomarkers in stroke have reached an outstanding and remarkable revision in the very recent years. Besides the diagnostic and prognostic role of some inflammatory markers, many further molecules and biological factors have been added to the list, including tissue derived cytokines, growth factor-like molecules, hormones, and microRNAs. The literatures on brain derived growth factor and other neuroimmune mediators, bone-skeletal muscle biomarkers, cellular and immunity biomarkers, and the role of microRNAs in stroke recovery were reviewed. To date, biomarkers represent a possible challenge in the diagnostic and prognostic evaluation of stroke onset, pathogenesis, and recovery. Many molecules are still under investigation and may become promising and encouraging biomarkers. Experimental and clinical research should increase this list and promote new discoveries in this field, to improve stroke diagnosis and treatment.
Collapse
Affiliation(s)
- Marialuisa Gandolfi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- UOC Neurorehabilitation, AOUI Verona, Verona, Italy
| | - Nicola Smania
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- UOC Neurorehabilitation, AOUI Verona, Verona, Italy
| | - Antonio Vella
- Immunology Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Alessandro Picelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- UOC Neurorehabilitation, AOUI Verona, Verona, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
13
|
Atorvastatin Modulates Regulatory T Cells and Attenuates Cerebral Damage in a Model of Transient Middle Cerebral Artery Occlusion in Rats. J Neuroimmune Pharmacol 2016; 12:152-162. [PMID: 27614888 DOI: 10.1007/s11481-016-9706-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/29/2016] [Indexed: 10/21/2022]
Abstract
Regulatory T cells (Tregs) inhibit the activation of the immune response which could down-regulate the systemic and focal activation observed during ischemic stroke. In fact, in animal models, Tregs infiltrate the infarcted brain and reduce the pro-inflammatory cytokine production and infarct volume, mainly in late stages of ischemia. Recently, an expansion and greater suppressive capacity of circulating Tregs after treatment with statins was observed, in addition to their cardio- and neuroprotective actions demonstrated previously. Thus, to determine whether Treg modulation mediated by statins can also be beneficial during stroke, cerebral ischemia was artificially induced in Wistar rats by transient middle cerebral artery occlusion (tMCAO) during 60 minutes with subsequent reperfusion for 7 days. Six hours after surgery, some animals were treated with atorvastatin (ATV, 10 mg/kg) or carboxymethylcellulose as vehicle at the same concentration every other day during 7 days. Some animals were sham operated as control group of surgery. Interestingly, ATV treatment prevented the development of infarct volume, reduced the neurological deficits, and the circulating and cervical lymph node CD25+FoxP3+ Treg population. Moreover, there was a reduction of glial cell activation, which correlated with decreased circulating Tregs. Remarkably, treatment with ATV induced an increase in the frequency of CD4+CD25+ T cells, in particular of those expressing CTLA-4, in brain samples. Together, these results suggest that ATV can modulate Tregs in peripheral tissue and favor their accumulation in the brain, where they can exert neuroprotective actions maybe by the reduction of glial cell activation.
Collapse
|
14
|
Zhang H. Progress in Research on the Mechanism and Treatment of Post-stroke Infection. INFECTION INTERNATIONAL 2016. [DOI: 10.1515/ii-2017-0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractPost-stroke infection hinders the recovery of stroke patients and can even cause death. The main mechanism of post-stroke infection is related with the post-stroke center, the activation of the peripheral immune system, and the release of inflammatory factors caused by the lesion area and pathophysiological changes in the body. Therefore, elucidating the body’s abnormal immune inflammatory responses after stroke is crucial for the prevention, treatment, and alleviation of post-stroke infection.
Collapse
|
15
|
Barnett-Vanes A, Sharrock A, Birrell MA, Rankin S. A Single 9-Colour Flow Cytometric Method to Characterise Major Leukocyte Populations in the Rat: Validation in a Model of LPS-Induced Pulmonary Inflammation. PLoS One 2016; 11:e0142520. [PMID: 26764486 PMCID: PMC4713146 DOI: 10.1371/journal.pone.0142520] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/22/2015] [Indexed: 01/08/2023] Open
Abstract
The rat is a commonly used model for immunological investigation. Yet basic research and characterisation of leukocyte populations and sub-sets lags far behind murine research, with inconsistency on reported leukocyte markers and their overlap. These shortcomings limit the opportunity for more complex and advanced rat immunology research. In this study, we developed a robust 9-colour flow-cytometric protocol to elucidate the major blood and tissue rat leukocyte populations, and validated it in a model of LPS-induced pulmonary inflammation. Blood and tissues (lung, BALF, spleen, liver, bone marrow) from naïve Sprague-Dawley rats were collected and analysed by flow cytometry (FCM). Rats were exposed to aerosolised saline or LPS (1 mg/mL), at 3 and 24 hrs thereafter blood, lung and BALF were collected and analysed using FCM and ELISA. Neutrophils, two monocyte subsets, NK Cells, B Cells, CD4+, CD8+ T Cells and alveolar macrophages can be identified simultaneously across different tissues using a 9-colour panel. Neutrophils and monocytes can be distinguished based upon differential expression of CD43 and His48. Neutrophils and CD43Lo/His48Hi monocyte-macrophages are elevated in the lung at 3 and 24 hrs during LPS-induced pulmonary inflammation. This validated method for leukocyte enumeration will offer a platform for greater consistency in future rat immunology and inflammation research.
Collapse
Affiliation(s)
- Ashton Barnett-Vanes
- Inflammation, Repair and Development Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Centre for Blast Injury Studies, Imperial College London, London, United Kingdom
| | - Anna Sharrock
- Inflammation, Repair and Development Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Centre for Blast Injury Studies, Imperial College London, London, United Kingdom
- Academic Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, United Kingdom
| | - Mark A. Birrell
- Respiratory Pharmacology Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sara Rankin
- Inflammation, Repair and Development Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Centre for Blast Injury Studies, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
16
|
The Role of Spleen-Derived Immune Cells in Ischemic Brain Injury. SPRINGER SERIES IN TRANSLATIONAL STROKE RESEARCH 2016. [DOI: 10.1007/978-3-319-32337-4_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Abstract
Stroke results in cerebral inflammation that causes brain injury and triggers immunodepression, resulting in an increased incidence of morbidity and mortality secondary to remote infection. It is well known that T cells modulate brain inflammation after ischemic stroke, and targeting T cells may be an innovative therapeutic strategy for stroke treatment. T cell deficiency is neuro-protective, but the observed protective effects differ between ischemic models. Recent studies suggest different T cell subsets may have distinct effects on the injured brain. In addition to their role in cerebral inflammation, T cells also play a role in stroke-induced immunodepression. Therefore, T cell-targeted therapies designed to provide protection against brain inflammation might paradoxically contribute to remote organ infection and mortality. Further investigations are required to determine the role of specific T cell subsets in cerebral inflammation and stroke-induced immunodepression, the optimal therapeutic window for treatment, and the appropriate dose of anti-T cell therapy.
Collapse
Affiliation(s)
- Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | | | | | | |
Collapse
|
18
|
Shi L, Qin J, Song B, Wang QM, Zhang R, Liu X, Liu Y, Hou H, Chen X, Ma X, Jiang C, Sun X, Gong G, Xu Y. Increased frequency of circulating regulatory T cells in patients with acute cerebral hemorrhage. Neurosci Lett 2015; 591:115-120. [PMID: 25703227 DOI: 10.1016/j.neulet.2015.02.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/01/2015] [Accepted: 02/17/2015] [Indexed: 01/09/2023]
Abstract
Cerebral hemorrhage (ICH) is a serious stroke subtype, currently lacking effective treatment. Recent research has shown that CD4(+)CD25(+)FOXP3(+) regulatory T cells (Tregs) play a key role in the immune response of ischemic stroke. However, Tregs in human hemorrhagic stroke are poorly investigated. In this study, a total of 90 ICH patients and 60 healthy controls were recruited. The frequency of circulating Tregs, plasma levels of TGF-β and IL-10, and the severity of neural dysfunction in ICH patients were investigated at different time points post ICH. We found that the peripheral frequency of Tregs in ICH patients was significantly increased, accompanied by boosted activated T cells. Importantly, the elevation of circulating Tregs in patients with severe dysfunction was much higher than that in less-severe patients, suggesting that disease severity affects circulating Tregs to exert regulatory function. Furthermore, both TGF-β and IL-10 that are related to the function of Tregs, were also increased in the peripheral blood of ICH patients. Our results demonstrate that Tregs-mediated immune imbalance might affect the development and severity of ICH, and suggest that Tregs may be used as tools and targets of cellular immunotherapy to effectively treat acute hemorrhagic stroke.
Collapse
Affiliation(s)
- Lijin Shi
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China; Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Henan Province 453100, China
| | - Jie Qin
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Bo Song
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Qing Mei Wang
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA 02114, United States
| | - Rui Zhang
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Xinjing Liu
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Yutao Liu
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Haiman Hou
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Xiulan Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Xinxiang Medical University, Henan Province 453100, China
| | - Xun Ma
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Chenyang Jiang
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Xiao Sun
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China
| | - Guangming Gong
- Department of Microbiology and Immunology, Basic Medical College of Zhengzhou University, Henan Province 450000, China.
| | - Yuming Xu
- Third Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan Province 450052, China.
| |
Collapse
|
19
|
Shen J, Zhu Y, Yu H, Fan ZX, Xiao F, Wu P, Zhang QH, Xiong XX, Pan JW, Zhan RY. Buyang Huanwu decoction increases angiopoietin-1 expression and promotes angiogenesis and functional outcome after focal cerebral ischemia. J Zhejiang Univ Sci B 2014; 15:272-80. [PMID: 24599691 DOI: 10.1631/jzus.b1300166] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Buyang Huanwu decoction (BYHWD), a traditional Chinese herbal prescription, has been widely used clinically to treat stroke in China for hundreds of years; however, the mechanisms of this drug for stroke treatment are still unclear. This study aims to observe the cerebral angiogenesis effects of BYHWD on chronic brain injury after focal cerebral ischemia in rats and to explore its possible mechanisms. The ischemia was induced by occlusion of the right middle cerebral artery for 90 min. BYHWD (12.5 and 25.0 g/(kg ∙ d), equivalent to the dry weight of the raw materials) was orally administered twice a day beginning 2 h after surgery. BYHWD significantly attenuated the neurological dysfunction, infarct volume, and brain atrophy after ischemia. There was a significant increase in the microvessel density, as assessed by immunofluorescence CD31, and a significant increase in angiopoietin-1 (Ang-1) in the penumbra areas of the rats was shown by immunohistochemical staining and Western blotting. The results indicate that the neurorestorative effects of BYHWD are associated with angiogenesis and the enhancement of the expressions of Ang-1 on chronic brain injury after focal cerebral ischemia.
Collapse
Affiliation(s)
- Jian Shen
- Brain Medicine Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China; Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China; Department of Anesthesiology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Xiong X, Xie R, Zhang H, Gu L, Xie W, Cheng M, Jian Z, Kovacina K, Zhao H. PRAS40 plays a pivotal role in protecting against stroke by linking the Akt and mTOR pathways. Neurobiol Dis 2014; 66:43-52. [PMID: 24583056 DOI: 10.1016/j.nbd.2014.02.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 01/08/2014] [Accepted: 02/19/2014] [Indexed: 11/16/2022] Open
Abstract
The proline-rich Akt substrate of 40kDa (PRAS40) protein is not only a substrate of the protein kinase Akt but also a component of the mTOR complex 1 (mTORC1), thus it links the Akt and the mTOR pathways. We investigated the potential protective role of PRAS40 in cerebral ischemia and its underlying mechanisms by using rats with lentiviral over-expression of PRAS40 and mice with PRAS40 gene knockout (PRAS40 KO). Our results show that gene transfer of PRAS40 reduced infarction size in rats by promoting phosphorylation of Akt, FKHR (FOXO1), PRAS40, and mTOR. In contrast, PRAS40 KO increased infarction size. Although the PRAS40 KO under normal condition did not alter baseline levels of phosphorylated proteins in the Akt and mTOR pathways, PRAS40 KO that underwent stroke exhibited reduced protein levels of p-S6K and p-S6 in the mTOR pathway but not p-Akt, or p-PTEN in the Akt pathway. Furthermore, co-immunoprecipitation suggests that there were less interactive effects between Akt and mTOR in the PRAS40 KO. In conclusion, PRAS40 appears to reduce brain injury by converting cell signaling from Akt to mTOR.
Collapse
Affiliation(s)
- Xiaoxing Xiong
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Rong Xie
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hongfei Zhang
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Lijuan Gu
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Weiying Xie
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Michelle Cheng
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Zhihong Jian
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Kristina Kovacina
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| | - Heng Zhao
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
21
|
Zhang YY, Li HX, Chen YY, Fang H, Yu YN, Liu J, Jing ZW, Wang Z, Wang YY. Convergent and divergent pathways decoding hierarchical additive mechanisms in treating cerebral ischemia-reperfusion injury. CNS Neurosci Ther 2013; 20:253-63. [PMID: 24351012 DOI: 10.1111/cns.12205] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Cerebral ischemia is considered to be a highly complex disease resulting from the complicated interplay of multiple pathways. Disappointedly, most of the previous studies were limited to a single gene or a single pathway. The extent to which all involved pathways are translated into fusing mechanisms of a combination therapy is of fundamental importance. AIMS We report an integrative strategy to reveal the additive mechanism that a combination (BJ) of compound baicalin (BA) and jasminoidin (JA) fights against cerebral ischemia based on variation of pathways and functional communities. RESULTS We identified six pathways of BJ group that shared diverse additive index from 0.09 to 1, which assembled broad cross talks from seven pathways of BA and 16 pathways of JA both at horizontal and vertical levels. Besides a total of 60 overlapping functions as a robust integration background among the three groups based on significantly differential subnetworks, additive mechanism with strong confidence by networks altered functions. CONCLUSIONS These results provide strong evidence that the additive mechanism is more complex than previously appreciated, and an integrative analysis of pathways may suggest an important paradigm for revealing pharmacological mechanisms underlying drug combinations.
Collapse
Affiliation(s)
- Ying-Ying Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Xiong XX, Gu LJ, Shen J, Kang XH, Zheng YY, Yue SB, Zhu SM. Probenecid protects against transient focal cerebral ischemic injury by inhibiting HMGB1 release and attenuating AQP4 expression in mice. Neurochem Res 2013; 39:216-24. [PMID: 24317635 DOI: 10.1007/s11064-013-1212-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 10/31/2013] [Accepted: 11/25/2013] [Indexed: 12/11/2022]
Abstract
Stroke results in inflammation, brain edema, and neuronal death. However, effective neuroprotectants are not available. Recent studies have shown that high mobility group box-1 (HMGB1), a proinflammatory cytokine, contributes to ischemic brain injury. Aquaporin 4 (AQP4), a water channel protein, is considered to play a pivotal role in ischemia-induced brain edema. More recently, studies have shown that pannexin 1 channels are involved in cerebral ischemic injury and the cellular inflammatory response. Here, we examined whether the pannexin 1 channel inhibitor probenecid could reduce focal ischemic brain injury by inhibiting cerebral inflammation and edema. Transient focal ischemia was induced in C57BL/6J mice by middle cerebral artery occlusion (MCAO) for 1 h. Infarct volume, neurological score and cerebral water content were evaluated 48 h after MCAO. Immunostaining, western blot analysis and ELISA were used to assess the effects of probenecid on the cellular inflammatory response, HMGB1 release and AQP4 expression. Administration of probenecid reduced infarct size, decreased cerebral water content, inhibited neuronal death, and reduced inflammation in the brain 48 h after stroke. In addition, HMGB1 release from neurons was significantly diminished and serum HMGB1 levels were substantially reduced following probenecid treatment. Moreover, AQP4 protein expression was downregulated in the cortical penumbra following post-stroke treatment with probenecid. These results suggest that probenecid, a powerful pannexin 1 channel inhibitor, protects against ischemic brain injury by inhibiting cerebral inflammation and edema.
Collapse
Affiliation(s)
- Xiao-Xing Xiong
- Department of Anesthesia, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Gu LJ, Xiong XX, Ito T, Lee J, Xu BH, Krams S, Steinberg GK, Zhao H. Moderate hypothermia inhibits brain inflammation and attenuates stroke-induced immunodepression in rats. CNS Neurosci Ther 2013; 20:67-75. [PMID: 23981596 DOI: 10.1111/cns.12160] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 07/14/2013] [Accepted: 07/15/2013] [Indexed: 01/07/2023] Open
Abstract
AIMS Stroke causes both brain inflammation and immunodepression. Mild-to-moderate hypothermia is known to attenuate brain inflammation, but its role in stroke-induced immunodepression (SIID) of the peripheral immune system remains unknown. This study investigated the effects in rats of moderate intra-ischemic hypothermia on SIID and brain inflammation. METHODS Stroke was induced in rats by permanent distal middle cerebral artery occlusion combined with transient bilateral common carotid artery occlusion, while body temperature was reduced to 30°C. Real-time PCR, flow cytometry, in vitro T-cell proliferation assays, in vivo delayed-type hypersensitivity (DTH) reaction and confocal microscopy were used to study SIID and brain inflammation. RESULTS Brief intra-ischemic hypothermia helped maintain certain leukocytes in the peripheral blood and spleen and enhanced T-cell proliferation in vitro and delayed-type hypersensitivity in vivo, suggesting that hypothermia reduces SIID. In contrast, in the brain, brief intra-Ischemic hypothermia inhibited mRNA expression of anti-inflammatory cytokine IL-10 and proinflammatory mediators INF-γ, TNF-α, IL-2, IL-1β and MIP-2. Brief intra-Ischemic hypothermia also attenuated the infiltration of lymphocytes, neutrophils (MPO(+) cells) and macrophages (CD68(+) cells) into the ischemic brain, suggesting that hypothermia inhibited brain inflammation. CONCLUSIONS Brief intra-ischemic hypothermia attenuated SIID and protected against acute brain inflammation.
Collapse
Affiliation(s)
- Li-Juan Gu
- Department of Neurosurgery, Stanford Stroke Center and Stanford Institute for Neuro-Innovation and Translational Neurosciences, Stanford University, Stanford, CA, USA; Department of Basic Medicine, Hangzhou Normal University, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|