1
|
Xu Y, Peng J, Yan Y, Gao M, Zang H, Cheng L, Zhou Y. CD19 + B cell depletion: a novel strategy to alleviate ischemic stroke damage. Front Immunol 2025; 16:1528471. [PMID: 40313936 PMCID: PMC12043492 DOI: 10.3389/fimmu.2025.1528471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Background Ischemic stroke, accounting for approximately 80% of all stroke cases, is a major public health challenge and a leading cause of death and disability worldwide. Current treatments primarily involve thrombolytic therapy, limited to a 4.5-hour window due to the risk of complications, underscoring the need for new therapeutic targets. Systemic inflammation plays a critical role in stroke progression, with immune cells infiltrating the brain and exacerbating damage. B cells, in particular, have been implicated in stroke pathogenesis, although their exact role remains contentious. This study examines anti-CD19 antibody (aCD19 Ab) treatment in a stroke model to determine if CD19+ B cell depletion can reduce infarct size and alleviate inflammation. Results This study investigated whether temporary inhibition of B-cell activity using an aCD19 Ab could alleviate ischemic brain injury in a stroke mouse model by regulating cerebral and systemic immune reactions. Mice subjected to middle cerebral artery occlusion (MCAO) exhibited significant reductions in infarct size and brain edema, prolonged post-MCAO survival, and improved behavioral outcomes following aCD19 Ab treatment. Transmission electron microscopy (TEM) and Computed Tomography Angiography (CTA) results revealed a reduction in microvascular endothelial edema, decreased mitochondrial damage in neurons, reduced neuronal apoptosis, and a favorable reconstruction of the cerebral vascular network. Additionally, B cell inhibition reduced pro-inflammatory cytokines and immune cells in the brain and peripheral circulation. The immune response alterations observed in the MCAO/R group were consistent with the trends indicated by stroke patient data. Conclusions Temporary inhibition of B-cell activity via aCD19 antibody injection alleviated ischemic brain injury in a mouse model of stroke by suppressing systemic immune reactions. Changes in immune cells within the meninges may play a role, and further investigation is needed to understand the mechanisms involved. These findings suggest that cerebral and systemic immune responses contribute to the pathogenesis of ischemic stroke, and temporary B cell depletion may represent a potential therapeutic target for stroke therapy.
Collapse
Affiliation(s)
- Yu Xu
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Jing Peng
- National Engineering Research Center of Human Stem Cell, Changsha, China
| | - Yizhong Yan
- National Engineering Research Center of Human Stem Cell, Changsha, China
| | - Min Gao
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - HongJing Zang
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lamei Cheng
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- National Engineering Research Center of Human Stem Cell, Changsha, China
- Hunan Guangxiu Hi-tech Life Technology Co. Ltd, Changsha, China
| | - Yu Zhou
- Department of Neurosurgery, The Second Xiangya Hospital of Central South University, Changsha, China
- National Engineering Research Center of Human Stem Cell, Changsha, China
- Hunan Guangxiu Hi-tech Life Technology Co. Ltd, Changsha, China
| |
Collapse
|
2
|
Zeng M, Peng M, Liang J, Sun H. The Role of Gut Microbiota in Blood-Brain Barrier Disruption after Stroke. Mol Neurobiol 2024; 61:9735-9755. [PMID: 37498481 DOI: 10.1007/s12035-023-03512-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Growing evidence has proved that alterations in the gut microbiota have been linked to neurological disorders including stroke. Structural and functional disruption of the blood-brain barrier (BBB) is observed after stroke. In this context, there is pioneering evidence supporting that gut microbiota may be involved in the pathogenesis of stroke by regulating the BBB function. However, only a few experimental studies have been performed on stroke models to observe the BBB by altering the structure of gut microbiota, which warrant further exploration. Therefore, in order to provide a novel mechanism for stroke and highlight new insights into BBB modification as a stroke intervention, this review summarizes existing evidence of the relationship between gut microbiota and BBB integrity and discusses the mechanisms of gut microbiota on BBB dysfunction and its role in stroke.
Collapse
Affiliation(s)
- Meiqin Zeng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Meichang Peng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jianhao Liang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Xu Z, Yang F, Zheng L. Uncovering the dual roles of peripheral immune cells and their connections to brain cells in stroke and post-stroke stages through single-cell sequencing. Front Neurosci 2024; 18:1443438. [PMID: 39633897 PMCID: PMC11614781 DOI: 10.3389/fnins.2024.1443438] [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: 06/04/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
Ischemic stroke is a cerebrovascular disease that affects the blood vessels and the blood supply to the brain, making it the second leading cause of death worldwide. Studies suggest that immune cells play a dual role during the inflammatory and recovery phases of stroke. However, in-depth investigations of specific cell subtypes and their differentiation trajectories remain to be elucidated. In this review, we highlight the application of single-cell RNA sequencing (scRNA-seq) for the unbiased identification of cell heterogeneity in brain and peripheral blood mononuclear cells (PBMCs) during and after a stroke. Our goal is to explore the phenotypic landscape of cells with different roles in this context. Specifically, we provide an overview of the roles, cell surface markers, immune cell-released cytokines, and intercellular interactions identified in major immune cells during and after stroke, as identified by different technologies. Additionally, we summarize the connection between immune cells in peripheral blood and the brain via their differentiation trajectories. By synthesizing the application of scRNA-seq in the combined analysis of PBMCs and brain tissue at higher sampling frequencies, we aim to unveil the dual role of peripheral immune cells, which could facilitate the development of new treatment strategies for ischemic stroke.
Collapse
Affiliation(s)
- Zheng Xu
- Department of Neurology, Southern University of Sciences and Technology Yantian Hospital, Shenzhen, China
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fan Yang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lifang Zheng
- Department of Neurology, Southern University of Sciences and Technology Yantian Hospital, Shenzhen, China
| |
Collapse
|
4
|
Xie L, He M, Ying C, Chu H. Mechanisms of inflammation after ischemic stroke in brain-peripheral crosstalk. Front Mol Neurosci 2024; 17:1400808. [PMID: 38932932 PMCID: PMC11199882 DOI: 10.3389/fnmol.2024.1400808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Stroke is a devastating disease with high morbidity, disability, and mortality, among which ischemic stroke is more common. However, there is still a lack of effective methods to improve the prognosis and reduce the incidence of its complications. At present, there is evidence that peripheral organs are involved in the inflammatory response after stroke. Moreover, the interaction between central and peripheral inflammation includes the activation of resident and peripheral immune cells, as well as the activation of inflammation-related signaling pathways, which all play an important role in the pathophysiology of stroke. In this review, we discuss the mechanisms of inflammatory response after ischemic stroke, as well as the interactions through circulatory pathways between peripheral organs (such as the gut, heart, lung and spleen) and the brain to mediate and regulate inflammation after ischemic stroke. We also propose the potential role of meningeal lymphatic vessels (MLVs)-cervical lymph nodes (CLNs) as a brain-peripheral crosstalk lymphatic pathway in ischemic stroke. In addition, we also summarize the mechanisms of anti-inflammatory drugs in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ling Xie
- Department of Critical Medicine, First People's Hospital of Linping District, Hangzhou, China
| | - Ming He
- Department of Critical Medicine, First People's Hospital of Linping District, Hangzhou, China
| | - Caidi Ying
- Department of Hepatobiliary and Pancreatic Surgery, The Traditional Chinese Medicine Hospital of Ningbo, Ningbo, China
| | - Haifeng Chu
- Department of Neurosurgery, The Traditional Chinese Medicine Hospital of Linping District, Hangzhou, China
| |
Collapse
|
5
|
Wu S, Tabassum S, Payne CT, Hu H, Gusdon AM, Choi HA, Ren XS. Updates of the role of B-cells in ischemic stroke. Front Cell Neurosci 2024; 18:1340756. [PMID: 38550918 PMCID: PMC10972894 DOI: 10.3389/fncel.2024.1340756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/27/2024] [Indexed: 10/11/2024] Open
Abstract
Ischemic stroke is a major disease causing death and disability in the elderly and is one of the major diseases that seriously threaten human health and cause a great economic burden. In the early stage of ischemic stroke, neuronal structure is destroyed, resulting in death or damage, and the release of a variety of damage-associated pattern molecules induces an increase in neuroglial activation, peripheral immune response, and secretion of inflammatory mediators, which further exacerbates the damage to the blood-brain barrier, exacerbates cerebral edema, and microcirculatory impairment, triggering secondary brain injuries. After the acute phase of stroke, various immune cells initiate a protective effect, which is released step by step and contributes to the repair of neuronal cells through phenotypic changes. In addition, ischemic stroke induces Central Nervous System (CNS) immunosuppression, and the interaction between the two influences the outcome of stroke. Therefore, modulating the immune response of the CNS to reduce the inflammatory response and immune damage during stroke is important for the protection of brain function and long-term recovery after stroke, and modulating the immune function of the CNS is expected to be a novel therapeutic strategy. However, there are fewer studies on B-cells in brain function protection, which may play a dual role in the stroke process, and the understanding of this cell is still incomplete. We review the existing studies on the mechanisms of the role of B-cells, inflammatory response, and immune response in the development of ischemic stroke and provide a reference for the development of adjuvant therapeutic drugs for ischemic stroke targeting inflammatory injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xuefang S. Ren
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, United States
| |
Collapse
|
6
|
Farag E, Machado S, Argalious M. Multiorgan talks in the presence of brain injury. Curr Opin Anaesthesiol 2023; 36:476-484. [PMID: 37552078 DOI: 10.1097/aco.0000000000001292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
PURPOSE OF REVIEW The brain is the command center of the rest of the body organs. The normal multiorgan talks between the brain and the rest of the body organs are essential for the normal body homeostasis. In the presence of brain injury, the disturbed talks between the brain and the rest of body organs will result in several pathological conditions. The aim of this review is to present the most recent findings for the pathological conditions that would result from the impaired multiorgan talks in the presence of brain injury. RECENT FINDINGS The brain injury such as in acute ischemic stroke, subarachnoid hemorrhage and traumatic brain injury will result in cascade of pathological talks between the brain and the rest of body organs. These pathological talks could result in pathological conditions such as cardiomyopathy, acute lung and kidney injuries, impaired liver functions, and impaired gut barrier permeability as well. SUMMARY Better understanding of the pathological conditions that could result from the impaired multiorgan talks in the presence of brain injury will open the doors for precise targeted therapies in the future for myriad of pathological conditions.
Collapse
Affiliation(s)
- Ehab Farag
- Department of General Anesthesiology, Anesthesia Institute, Cleveland Clinic, Ohio, USA
| | | | | |
Collapse
|
7
|
Liu S, Zhang Z, He Y, Kong L, Jin Q, Qi X, Qi D, Gao Y. Inhibiting leukocyte-endothelial cell interactions by Chinese medicine Tongxinluo capsule alleviates no-reflow after arterial recanalization in ischemic stroke. CNS Neurosci Ther 2023; 29:3014-3030. [PMID: 37122157 PMCID: PMC10493667 DOI: 10.1111/cns.14242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 05/02/2023] Open
Abstract
AIMS Despite successful vascular recanalization in stroke, one-fourth of patients have an unfavorable outcome due to no-reflow. The pathogenesis of no-reflow is fully unclear, and therapeutic strategies are lacking. Upon traditional Chinese medicine, Tongxinluo capsule (TXL) is a potential therapeutic agent for no-reflow. Thus, this study is aimed to investigate the pathogenesis of no-reflow in stroke, and whether TXL could alleviate no-reflow as well as its potential mechanisms of action. METHODS Mice were orally administered with TXL (3.0 g/kg/d) after transient middle cerebral artery occlusion. We examined the following parameters: neurological function, no-reflow, leukocyte-endothelial cell interactions, HE staining, leukocyte subtypes, adhesion molecules, and chemokines. RESULTS Our results showed stroke caused neurological deficits, neuron death, and no-reflow. Adherent and aggregated leukocytes obstructed microvessels as well as leukocyte infiltration in ischemic brain. Leukocyte subtypes changed after stroke mainly including neutrophils, lymphocytes, regulatory T cells, suppressor T cells, helper T type 1 (Th1) cells, Th2 cells, B cells, macrophages, natural killer cells, and dendritic cells. Stroke resulted in upregulated expression of adhesion molecules (P-selectin, E-selectin, and ICAM-1) and chemokines (CC-chemokine ligand (CCL)-2, CCL-3, CCL-4, CCL-5, and chemokine C-X-C ligand 1 (CXCL-1)). Notably, TXL improved neurological deficits, protected neurons, alleviated no-reflow and leukocyte-endothelial cell interactions, regulated multiple leukocyte subtypes, and inhibited the expression of various inflammatory mediators. CONCLUSION Leukocyte-endothelial cell interactions mediated by multiple inflammatory factors are an important cause of no-reflow in stroke. Accordingly, TXL could alleviate no-reflow via suppressing the interactions through modulating various leukocyte subtypes and inhibiting the expression of multiple inflammatory mediators.
Collapse
Affiliation(s)
- Shen Liu
- Department of Neurology of TCM, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
| | - Zhaoxu Zhang
- Department of NeurologyPeking University People's HospitalBeijingChina
| | - Yannan He
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
| | - Lingbo Kong
- Department of Neurology of TCM, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
| | - Qiushuo Jin
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
| | - Xiangjia Qi
- Department of NeurologyThe First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan HospitalJinanChina
| | - Dahe Qi
- Department of Neurology of TCM, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
| | - Ying Gao
- Department of Neurology of TCM, Dongzhimen HospitalBeijing University of Chinese MedicineBeijingChina
- Institute for Brain Disorders, Beijing University of Chinese MedicineBeijingChina
| |
Collapse
|
8
|
Nguyen JN, Chauhan A. Bystanders or not? Microglia and lymphocytes in aging and stroke. Neural Regen Res 2023; 18:1397-1403. [PMID: 36571333 PMCID: PMC10075112 DOI: 10.4103/1673-5374.360345] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
As the average age of the world population increases, more people will face debilitating aging-associated conditions, including dementia and stroke. Not only does the incidence of these conditions increase with age, but the recovery afterward is often worse in older patients. Researchers and health professionals must unveil and understand the factors behind age-associated diseases to develop a therapy for older patients. Aging causes profound changes in the immune system including the activation of microglia in the brain. Activated microglia promote T lymphocyte transmigration leading to an increase in neuroinflammation, white matter damage, and cognitive impairment in both older humans and rodents. The presence of T and B lymphocytes is observed in the aged brain and correlates with worse stroke outcomes. Preclinical strategies in stroke target either microglia or the lymphocytes or the communications between them to promote functional recovery in aged subjects. In this review, we examine the role of the microglia and T and B lymphocytes in aging and how they contribute to cognitive impairment. Additionally, we provide an important update on the contribution of these cells and their interactions in preclinical aged stroke.
Collapse
Affiliation(s)
- Justin N. Nguyen
- University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Anjali Chauhan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| |
Collapse
|
9
|
Yamada H, Kase Y, Okano Y, Kim D, Goto M, Takahashi S, Okano H, Toda M. Subarachnoid hemorrhage triggers neuroinflammation of the entire cerebral cortex, leading to neuronal cell death. Inflamm Regen 2022; 42:61. [PMID: 36514181 DOI: 10.1186/s41232-022-00236-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/09/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a fatal disease, with early brain injury (EBI) occurring within 72 h of SAH injury contributes to its poor prognosis. EBI is a complicated phenomenon involving multiple mechanisms. Although neuroinflammation has been shown to be important prognosis factor of EBI, whether neuroinflammation spreads throughout the cerebrum and the extent of its depth in the cerebral cortex remain unknown. Knowing how inflammation spreads throughout the cerebrum is also important to determine if anti-inflammatory agents are a future therapeutic strategy for EBI. METHODS In this study, we induced SAH in mice by injecting hematoma into prechiasmatic cistern and created models of mild to severe SAH. In sections of the mouse cerebrum, we investigated neuroinflammation and neuronal cell death in the cortex distal to the hematoma injection site, from anterior to posterior region 24 h after SAH injury. RESULTS Neuroinflammation caused by SAH spread to all layers of the cerebral cortex from the anterior to the posterior part of the cerebrum via the invasion of activated microglia, and neuronal cell death increased in correlation with neuroinflammation. This trend increased with the severity of the disease. CONCLUSIONS Neuroinflammation caused by SAH had spread throughout the cerebrum, causing neuronal cell death. Considering that the cerebral cortex is responsible for long-term memory and movement, suppressing neuroinflammation in all layers of the cerebral cortex may improve the prognosis of patients with SAH.
Collapse
Affiliation(s)
- Hiroki Yamada
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yoshitaka Kase
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yuji Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Doyoon Kim
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Maraku Goto
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.,The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Satoshi Takahashi
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
10
|
Zhang Z, Lv M, Zhou X, Cui Y. Roles of peripheral immune cells in the recovery of neurological function after ischemic stroke. Front Cell Neurosci 2022; 16:1013905. [PMID: 36339825 PMCID: PMC9634819 DOI: 10.3389/fncel.2022.1013905] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/03/2022] [Indexed: 10/15/2023] Open
Abstract
Stroke is a leading cause of mortality and long-term disability worldwide, with limited spontaneous repair processes occurring after injury. Immune cells are involved in multiple aspects of ischemic stroke, from early damage processes to late recovery-related events. Compared with the substantial advances that have been made in elucidating how immune cells modulate acute ischemic injury, the understanding of the impact of the immune system on functional recovery is limited. In this review, we summarized the mechanisms of brain repair after ischemic stroke from both the neuronal and non-neuronal perspectives, and we review advances in understanding of the effects on functional recovery after ischemic stroke mediated by infiltrated peripheral innate and adaptive immune cells, immune cell-released cytokines and cell-cell interactions. We also highlight studies that advance our understanding of the mechanisms underlying functional recovery mediated by peripheral immune cells after ischemia. Insights into these processes will shed light on the double-edged role of infiltrated peripheral immune cells in functional recovery after ischemic stroke and provide clues for new therapies for improving neurological function.
Collapse
Affiliation(s)
- Zhaolong Zhang
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Mengfei Lv
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Xin Zhou
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Yu Cui
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
11
|
Zhu L, Huang L, Le A, Wang TJ, Zhang J, Chen X, Wang J, Wang J, Jiang C. Interactions between the Autonomic Nervous System and the Immune System after Stroke. Compr Physiol 2022; 12:3665-3704. [PMID: 35766834 DOI: 10.1002/cphy.c210047] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute stroke is one of the leading causes of morbidity and mortality worldwide. Stroke-induced immune-inflammatory response occurs in the perilesion areas and the periphery. Although stroke-induced immunosuppression may alleviate brain injury, it hinders brain repair as the immune-inflammatory response plays a bidirectional role after acute stroke. Furthermore, suppression of the systemic immune-inflammatory response increases the risk of life-threatening systemic bacterial infections after acute stroke. Therefore, it is essential to explore the mechanisms that underlie the stroke-induced immune-inflammatory response. Autonomic nervous system (ANS) activation is critical for regulating the local and systemic immune-inflammatory responses and may influence the prognosis of acute stroke. We review the changes in the sympathetic and parasympathetic nervous systems and their influence on the immune-inflammatory response after stroke. Importantly, this article summarizes the mechanisms on how ANS regulates the immune-inflammatory response through neurotransmitters and their receptors in immunocytes and immune organs after stroke. To facilitate translational research, we also discuss the promising therapeutic approaches modulating the activation of the ANS or the immune-inflammatory response to promote neurologic recovery after stroke. © 2022 American Physiological Society. Compr Physiol 12:3665-3704, 2022.
Collapse
Affiliation(s)
- Li Zhu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Anh Le
- Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Tom J Wang
- Winston Churchill High School, Potomac, Maryland, USA
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xuemei Chen
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Jian Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China.,Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
12
|
Liu R, Song P, Gu X, Liang W, Sun W, Hua Q, Zhang Y, Qiu Z. Comprehensive Landscape of Immune Infiltration and Aberrant Pathway Activation in Ischemic Stroke. Front Immunol 2022; 12:766724. [PMID: 35140708 PMCID: PMC8818702 DOI: 10.3389/fimmu.2021.766724] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Ischemic stroke (IS) is a multifactorial disease caused by the interaction of multiple environmental and genetic risk factors, and it is the most common cause of disability. The immune microenvironment and inflammatory response participate in the whole process of IS occurrence and development. Therefore, the rational use of relevant markers or characteristic pathways in the immune microenvironment will become one of the important therapeutic strategies for the treatment of IS. We collected peripheral blood samples from 10 patients diagnosed with IS at the First Affiliated Hospital of Gannan Medical University and First Affiliated Hospital, Jinan" University, and from 10 normal people. The GSE16561 dataset was downloaded from the Gene Expression Omnibus (GEO) database. xCell, gene set enrichment analysis (GSEA), single-sample GSEA (ssGSEA) and immune-related gene analysis were used to evaluate the differences in the immune microenvironment and characteristic pathways between the IS and control groups of the two datasets. xCell analysis showed that the IS-24h group had significantly reduced central memory CD8+ T cell, effector memory CD8+ T cell, B cell and Th1 cell scores and significantly increased M1 macrophage and macrophage scores. GSEA showed that the IS-24h group had significantly increased inflammation-related pathway activity(myeloid leukocyte activation, positive regulation of tumor necrosis factor biosynthetic process, myeloid leukocyte migration and leukocyte chemotaxis), platelet-related pathway activity(platelet activation, signaling and aggregation; protein polymerization; platelet degranulation; cell-cell contact zone) and pathology-related pathway activity (ERBB signaling pathway, positive regulation of ERK1 and ERK2 cascade, vascular endothelial growth factor receptor signaling pathway, and regulation of MAP kinase activity). Immune-related signature analysis showed that the macrophage signature, antigen presentation-related signature, cytotoxicity-related signature, B cell-related signature and inflammation-related signature were significantly lower in the IS-24h group than in the control group. In this study, we found that there were significant differences in the immune microenvironment between the peripheral blood of IS patients and control patients, as shown by the IS group having significantly reduced CD8+ Tcm, CD8+ Tem, B cell and Th1 cell scores and significantly increased macrophage and M1 macrophage scores. Additionally, inflammation-related, pathological, and platelet-related pathway activities were significantly higher in the IS group than in the control group.
Collapse
Affiliation(s)
- Rongrong Liu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Neurology, Ganzhou People’s Hospital, Ganzhou, China
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Pingping Song
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Xunhu Gu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weidong Liang
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Wei Sun
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Qian Hua
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yusheng Zhang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
- *Correspondence: Yusheng Zhang, ; Zhengang Qiu,
| | - Zhengang Qiu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Yusheng Zhang, ; Zhengang Qiu,
| |
Collapse
|
13
|
Tröscher AR, Gruber J, Wagner JN, Böhm V, Wahl AS, von Oertzen TJ. Inflammation Mediated Epileptogenesis as Possible Mechanism Underlying Ischemic Post-stroke Epilepsy. Front Aging Neurosci 2021; 13:781174. [PMID: 34966269 PMCID: PMC8711648 DOI: 10.3389/fnagi.2021.781174] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/23/2021] [Indexed: 01/19/2023] Open
Abstract
Post-stroke Epilepsy (PSE) is one of the most common forms of acquired epilepsy, especially in the elderly population. As people get increasingly older, the number of stroke patients is expected to rise and concomitantly the number of people with PSE. Although many patients are affected by post-ischemic epileptogenesis, not much is known about the underlying pathomechanisms resulting in the development of chronic seizures. A common hypothesis is that persistent neuroinflammation and glial scar formation cause aberrant neuronal firing. Here, we summarize the clinical features of PSE and describe in detail the inflammatory changes after an ischemic stroke as well as the chronic changes reported in epilepsy. Moreover, we discuss alterations and disturbances in blood-brain-barrier leakage, astrogliosis, and extracellular matrix changes in both, stroke and epilepsy. In the end, we provide an overview of commonalities of inflammatory reactions and cellular processes in the post-ischemic environment and epileptic brain and discuss how these research questions should be addressed in the future.
Collapse
Affiliation(s)
| | - Joachim Gruber
- Neurology I, Neuromed Campus, Kepler Universitätsklinikum, Linz, Austria.,Medical Faculty, Johannes Kepler University, Linz, Austria
| | - Judith N Wagner
- Neurology I, Neuromed Campus, Kepler Universitätsklinikum, Linz, Austria.,Medical Faculty, Johannes Kepler University, Linz, Austria
| | - Vincent Böhm
- Neurology I, Neuromed Campus, Kepler Universitätsklinikum, Linz, Austria.,Medical Faculty, Johannes Kepler University, Linz, Austria
| | - Anna-Sophia Wahl
- Brain Research Institute, University of Zurich, Zurich, Switzerland.,Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany
| | - Tim J von Oertzen
- Neurology I, Neuromed Campus, Kepler Universitätsklinikum, Linz, Austria.,Medical Faculty, Johannes Kepler University, Linz, Austria
| |
Collapse
|
14
|
Qi X, Lin H, Hou Y, Su X, Gao Y. Comprehensive Analysis of Potential miRNA-Target mRNA-Immunocyte Subtype Network in Cerebral Infarction. Eur Neurol 2021; 85:148-161. [PMID: 34544080 DOI: 10.1159/000518893] [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: 04/21/2021] [Accepted: 07/27/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Cerebral infarction (CI) is one of the leading causes of serious long-term disability and mortality. OBJECTIVE We aimed to identify potential miRNAs and target mRNAs and assess the involvement of immunocyte infiltration in the process of CI. METHODS First, miRNA and mRNA data were downloaded from the Gene Expression Omnibus database, followed by differential expression analysis. Second, correlation analysis between differentially expressed mRNAs and differential immunocyte subtypes was performed through the CIBERSORT algorithm. Third, the regulatory network between miRNAs and immunocyte subtype-related mRNAs was constructed followed by the functional analysis of these target mRNAs. Fourth, correlation validation between differentially expressed mRNAs and differential immunocyte subtypes was performed in the GSE37587 dataset. Finally, the diagnostic ability of immunocyte subtype-related mRNAs was tested. RESULTS Up to 17 differentially expressed miRNAs and 3,267 differentially expressed mRNAs were identified, among which 310 differentially expressed mRNAs were significantly associated with immunocyte subtypes. Several miRNA-target mRNA-immunocyte subtype networks including hsa-miR-671-3p-ZC3HC1-neutrophils, hsa-miR-625-CD5-monocytes, hsa-miR-122-ACOX1/DUSP1/NEDD9-neutrophils, hsa-miR-455-5p-SLC24A4-monocytes, and hsa-miR-455-5p-SORL1-neutrophils were identified. LAT, ACOX1, DUSP1, NEDD9, ZC3HC1, BIN1, AKT1, DNMT1, SLC24A4, and SORL1 had a potential diagnostic value for CI. CONCLUSIONS The network including miRNA, target mRNA, and immunocyte subtype may be novel regulators and diagnostic and therapeutic targets in CI.
Collapse
Affiliation(s)
- Xiuyan Qi
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Huiqian Lin
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Yongge Hou
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xiaohui Su
- Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Yanfang Gao
- Clinical Laboratory, Hebei Red Cross Boai Hospital, Shijiazhuang, China
| |
Collapse
|
15
|
Zou H, Zhou N, Huang Y, Luo A, Sun J. Phenotypes, roles, and modulation of regulatory lymphocytes in periodontitis and its associated systemic diseases. J Leukoc Biol 2021; 111:451-467. [PMID: 33884656 DOI: 10.1002/jlb.3vmr0321-027rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Periodontitis is a common chronic inflammatory disease that can result in tooth loss and poses a risk to systemic health. Lymphocytes play important roles in periodontitis through multiple mechanisms. Regulatory lymphocytes including regulatory B cells (Bregs) and T cells (Tregs) are the main immunosuppressive cells that maintain immune homeostasis, and are critical to our understanding of the pathogenesis of periodontitis and the development of effective treatments. In this review, we discuss the phenotypes, roles, and modulating strategies of regulatory lymphocytes including Bregs and Tregs in periodontitis and frequently cooccurring inflammatory diseases such as rheumatoid arthritis, Alzheimer disease, diabetes mellitus, and stroke. The current evidence suggests that restoring immune balance through therapeutic targeting of regulatory lymphocytes is a promising strategy for the treatment of periodontitis and other systemic inflammatory diseases.
Collapse
Affiliation(s)
- Hang Zou
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Niu Zhou
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guangzhou Zoo, Guangzhou, China
| | - Yilian Huang
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou, China
| | - Aoxiang Luo
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jianbo Sun
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
16
|
Qin X, Akter F, Qin L, Cheng J, Guo M, Yao S, Jian Z, Liu R, Wu S. Adaptive Immunity Regulation and Cerebral Ischemia. Front Immunol 2020; 11:689. [PMID: 32477327 PMCID: PMC7235404 DOI: 10.3389/fimmu.2020.00689] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 03/26/2020] [Indexed: 12/22/2022] Open
Abstract
Stroke is a disease that occurs due to a sudden interruption of the blood supply to the brain. It is a leading cause of death and disability worldwide. It is well-known that the immune system drives brain injury following an episode of ischemic stroke. The innate system and the adaptive system play distinct but synergistic roles following ischemia. The innate system can be activated by damage-associated molecular patterns (DAMPs), which are released from cells in the ischemic region. Damaged cells also release various other mediators that serve to increase inflammation and compromise the integrity of the blood–brain barrier (BBB). Within 24 h of an ischemic insult, the adaptive immune system is activated. This involves T cell and B cell-mediated inflammatory and humoral effects. These cells also stimulate the release of various interleukins and cytokines, which can modulate the inflammatory response. The adaptive immune system has been shown to contribute to a state of immunodepression following an ischemic episode, and this can increase the risk of infections. However, this phenomenon is equally important in preventing autoimmunity of the body to brain antigens that are released into the peripheral system as a result of BBB compromise. In this review, we highlight the key components of the adaptive immune system that are activated following cerebral ischemia.
Collapse
Affiliation(s)
- Xingping Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, United States
| | - Farhana Akter
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, United States.,Faculty of Arts and Sciences, Harvard University, Cambridge, MA, United States
| | - Lingxia Qin
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jing Cheng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mei Guo
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, United States
| | - Shun Yao
- Department of Neurosurgery, Center for Pituitary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Neurosurgery, Center for Skull Base and Pituitary Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Renzhong Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Songlin Wu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Courties G, Frodermann V, Honold L, Zheng Y, Herisson F, Schloss MJ, Sun Y, Presumey J, Severe N, Engblom C, Hulsmans M, Cremer S, Rohde D, Pittet MJ, Scadden DT, Swirski FK, Kim DE, Moskowitz MA, Nahrendorf M. Glucocorticoids Regulate Bone Marrow B Lymphopoiesis After Stroke. Circ Res 2020; 124:1372-1385. [PMID: 30782088 DOI: 10.1161/circresaha.118.314518] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE After a stroke, patients frequently experience altered systemic immunity resulting in peripheral immunosuppression and higher susceptibility to infections, which is at least partly attributed to lymphopenia. The mechanisms that profoundly change the systemic leukocyte repertoire after stroke are incompletely understood. Emerging evidence indicates that stroke alters hematopoietic output of the bone marrow. OBJECTIVE To explore the mechanisms that lead to defects of B lymphopoiesis after ischemic stroke. METHODS AND RESULTS We here report that ischemic stroke triggers brain-bone marrow communication via hormonal long-range signals that regulate hematopoietic B lineage decisions. Bone marrow fluorescence-activated cell sorter analyses and serial intravital microscopy indicate that transient middle cerebral artery occlusion in mice arrests B-cell development beginning at the pro-B-cell stage. This phenotype was not rescued in Myd88-/- and TLR4-/- mice with disrupted TLR (Toll-like receptor) signaling or after blockage of peripheral sympathetic nerves. Mechanistically, we identified stroke-induced glucocorticoid release as the main instigator of B lymphopoiesis defects. B-cell lineage-specific deletion of the GR (glucocorticoid receptor) in CD19-Cre loxP Nr3c1 mice attenuated lymphocytopenia after transient middle cerebral artery. In 20 patients with acute stroke, increased cortisol levels inversely correlated with blood lymphocyte numbers. CONCLUSIONS Our data demonstrate that the hypothalamic-pituitary-adrenal axis mediates B lymphopoiesis defects after ischemic stroke.
Collapse
Affiliation(s)
- Gabriel Courties
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Vanessa Frodermann
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Lisa Honold
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Yi Zheng
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Fanny Herisson
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Maximilian J Schloss
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Yuan Sun
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Jessy Presumey
- Massachusetts General Hospital and Program in Cellular and Molecular Medicine, Boston Children's Hospital and Department of Pediatrics (J.P.), Harvard Medical School, Boston
| | - Nicolas Severe
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.).,Harvard Stem Cell Institute, Cambridge, MA (N.S., D.T.S.).,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (N.S., D.T.S.)
| | - Camilla Engblom
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Maarten Hulsmans
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Sebastian Cremer
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - David Rohde
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Mikael J Pittet
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston (N.S., D.T.S.).,Harvard Stem Cell Institute, Cambridge, MA (N.S., D.T.S.).,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA (N.S., D.T.S.)
| | - Filip K Swirski
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston
| | - Dong-Eog Kim
- Molecular Imaging and Neurovascular Research Laboratory, Department of Neurology, Dongguk University College of Medicine, Goyang, South Korea (D.-E.K.)
| | - Michael A Moskowitz
- Stroke and Neurovascular Regulation Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown (M.A.M.)
| | - Matthias Nahrendorf
- From the Center for Systems Biology and Radiology Department (G.C., V.F., L.H., F.H., M.J.S., Y.S., C.E., M.H., S.C., D.R., M.J.P., F.K.S., M.N.), Harvard Medical School, Boston.,Cardiovascular Research Center (M.N.), Harvard Medical School, Boston
| |
Collapse
|
18
|
Zhao Y, Zhu T, Li H, Zhao J, Li X. Transplantation of Lymphocytes Co-Cultured with Human Cord Blood-Derived Multipotent Stem Cells Attenuates Inflammasome Activity in Ischemic Stroke. Clin Interv Aging 2019; 14:2261-2271. [PMID: 31908436 PMCID: PMC6927491 DOI: 10.2147/cia.s223595] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/11/2019] [Indexed: 01/25/2023] Open
Abstract
Background Manipulating the immune inflammatory response after cerebral ischemia has been a novel therapeutic strategy for ischemic stroke. This study attempted to investigate the effects of the transplantation of lymphocytes co-cultured with human cord blood-derived multipotent stem cells (HCB-SCs) on the inflammatory response in transient middle cerebral occlusion (tMCAO) rats. Methods The tMCAO rats were subjected to the transplantation of lymphocytes co-cultured with HCB-SCs through tail vein injection. Infarct size and neurological deficits were measured at 48 hrs after stroke. Neurological deficits were assessed using Bederson’s scoring system and tape removal test. Blood T cell flow cytometry was performed to measure the differentiation of regulatory T cells (Tregs). Western blot was used to detect the protein levels of inflammation-related molecules, apoptosis-related molecule, and signaling molecules in ischemic brain. TUNEL staining was performed to analyze cell apoptosis in ischemic cerebral cortex. Results The transplantation of lymphocytes co-cultured with HCB-SCs significantly improved the neurological defects, reduced ischemic brain damage, and increased the proportion of peripheral CD4+CD25+Foxp3+ Tregs. Meanwhile, the transplantation of co-cultured cells decreased the expression of NLRP3 inflammasome and associated factors, such as caspase-1 and IL-1β, and inhibited the activation of NF-κB, ERK and caspase-3 in ischemic brain. The co-cultured cells significantly decreased the number of tMCAO-induced cell apoptosis. Conclusion Lymphocytes co-cultured with HCB-SCs exhibit a neuroprotective effect after ischemic stroke by promoting Tregs differentiation and suppressing NLRP3 inflammasome activation and neuron apoptosis, and might be a promising therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Yanxin Zhao
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Tianrui Zhu
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Heng Li
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Jing Zhao
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| | - Xiaohong Li
- Department of Neurology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, People's Republic of China
| |
Collapse
|
19
|
Anitua E, Piñas L, Alkhraisat MH. Histopathological features of oral lichen planus and its response to corticosteroid therapy: A retrospective study. Medicine (Baltimore) 2019; 98:e18321. [PMID: 31860983 PMCID: PMC6940162 DOI: 10.1097/md.0000000000018321] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Oral lichen planus (OLP) exhibits variations in severity and response to corticosteroid therapy. This study aims to assess the histopathological features of OLP at the time of diagnosis and their relationship in response to corticosteroid therapy.In this retrospective study, OLP patients were selected if a histopathological report was available. Data were collected regarding patients' demographics and medical history. Clinical and histological data were also obtained. The outcomes were histopathological findings, clinical form of OLP, number of exacerbations per year, and the response to corticosteroid therapy.In this study, 100 OLP patients were enrolled. Basal layer hydropic degeneration and band-like subepithelial lymphocytes infiltrate were observed in all patients. Plasma cells, identified in 62% of OLP patients, were significantly associated with fewer disease exacerbations and better response to corticosteroid treatment.Identifying histopathological features that may affect the clinical course would be clinically helpful in tailoring patient management.
Collapse
Affiliation(s)
- Eduardo Anitua
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua)
- BTI Biotechnology Institute, Vitoria, Spain
| | - Laura Piñas
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua)
| | - Mohammad H. Alkhraisat
- University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua)
- BTI Biotechnology Institute, Vitoria, Spain
| |
Collapse
|
20
|
Wang Y, Zhang JH, Sheng J, Shao A. Immunoreactive Cells After Cerebral Ischemia. Front Immunol 2019; 10:2781. [PMID: 31849964 PMCID: PMC6902047 DOI: 10.3389/fimmu.2019.02781] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
The immune system is rapidly activated after ischemic stroke. As immune cells migrate and infiltrate across the blood-brain barrier into the ischemic region, a cascade of cellular and molecular biological reactions occur, involving migrated immune cells, resident glial cells, and the vascular endothelium. These events regulate infarction evolution and thus influence the outcome of ischemic stroke. Most immune cells exert dual effects on cerebral ischemia, and some crucial cells may become central targets in ischemic stroke treatment and rehabilitation.
Collapse
Affiliation(s)
- Yijie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jifang Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
21
|
Li XQ, Wang YY, Yang TT, Qian YN, Yin H, Zhong SS, A R, He Y, Xu BL, Liu GZ. Increased Peripheral CD137 Expression in a Mouse Model of Permanent Focal Cerebral Ischemia. Cell Mol Neurobiol 2019; 39:451-460. [PMID: 30778712 PMCID: PMC11469804 DOI: 10.1007/s10571-019-00661-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 02/07/2019] [Indexed: 11/30/2022]
Abstract
Various studies demonstrate that CD137 (TNFRSF9, 4-1BB) promotes atherosclerosis and vascular inflammation in experimental models via interactions with the CD137 ligand (CD137L). However, the exact role of CD137 in ischemic stroke remains unclear. In this study, we analyzed dynamic changes of peripheral CD137 expression on T cells in a mouse model of cerebral ischemia-middle cerebral artery occlusion (MCAO), as well as alternation of neurological function, infarct size and cerebral inflammatory status after inhibition of the CD137/CD137L pathway using an anti-CD137L monoclonal antibody. MCAO mice showed elevated surface expression of CD137 on T cells in both peripheral blood and lymphoid tissues during early cerebral ischemia. Remarkably, blockade of the CD137/CD137L pathway reduced the post-ischemic brain damage. Our findings indicate that enhanced CD137 costimulation occurs in early cerebral ischemia and promotes T cell activation, which in turn upregulates inflammatory immune response and possibly exerting deleterious effects on cerebral ischemia.
Collapse
Affiliation(s)
- Xiao-Qing Li
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yang-Yang Wang
- Department of Neurology, Peking University People's Hospital, Beijing, 100044, China
| | - Ting-Ting Yang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yi-Ning Qian
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - He Yin
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Shan-Shan Zhong
- Department of Neurology, Peking University People's Hospital, Beijing, 100044, China
| | - Rong A
- Department of Neurology, Peking University People's Hospital, Beijing, 100044, China
| | - Yang He
- Department of Neurology, Peking University People's Hospital, Beijing, 100044, China
| | - Bao-Lei Xu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Guang-Zhi Liu
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
22
|
Huang Q, Zhong W, Hu Z, Tang X. A review of the role of cav-1 in neuropathology and neural recovery after ischemic stroke. J Neuroinflammation 2018; 15:348. [PMID: 30572925 PMCID: PMC6302517 DOI: 10.1186/s12974-018-1387-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke starts a series of pathophysiological processes that cause brain injury. Caveolin-1 (cav-1) is an integrated protein and locates at the caveolar membrane. It has been demonstrated that cav-1 can protect blood–brain barrier (BBB) integrity by inhibiting matrix metalloproteases (MMPs) which degrade tight junction proteins. This article reviews recent developments in understanding the mechanisms underlying BBB dysfunction, neuroinflammation, and oxidative stress after ischemic stroke, and focuses on how cav-1 modulates a series of activities after ischemic stroke. In general, cav-1 reduces BBB permeability mainly by downregulating MMP9, reduces neuroinflammation through influencing cytokines and inflammatory cells, promotes nerve regeneration and angiogenesis via cav-1/VEGF pathway, reduces apoptosis, and reduces the damage mediated by oxidative stress. In addition, we also summarize some experimental results that are contrary to the above and explore possible reasons for these differences.
Collapse
Affiliation(s)
- Qianyi Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Wei Zhong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China.
| |
Collapse
|
23
|
Malone K, Amu S, Moore AC, Waeber C. The immune system and stroke: from current targets to future therapy. Immunol Cell Biol 2018; 97:5-16. [DOI: 10.1111/imcb.12191] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/11/2018] [Accepted: 07/16/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| | - Sylvie Amu
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| | - Anne C Moore
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics; School of Pharmacy; University College Cork; Cork Ireland
| |
Collapse
|
24
|
Duris K, Lipkova J, Splichal Z, Madaraszova T, Jurajda M. Early Inflammatory Response in the Brain and Anesthesia Recovery Time Evaluation After Experimental Subarachnoid Hemorrhage. Transl Stroke Res 2018; 10:10.1007/s12975-018-0641-z. [PMID: 29926382 DOI: 10.1007/s12975-018-0641-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 06/09/2018] [Accepted: 06/12/2018] [Indexed: 01/05/2023]
Abstract
The main objective was to evaluate, whether the subarachnoid hemorrhage (SAH)-associated early inflammatory response has focal or global character, i.e., whether areas distant to hematoma may be affected by an early inflammatory response. The second objective was to evaluate the association of anesthesia recovery time for basic reflexes/neurological functions with severity of SAH. SAH was induced in rats using an endovascular perforation model. Anesthesia recovery time was evaluated for pain reaction recovery time (spinal level), spontaneous ventilation recovery time (brain stem level), and consciousness recovery time (neocortical level). mRNA expressions of TNFα, IL-1β, IL-6, ICAM-1, and VCAM-1 in areas adjacent and distant to hematoma were evaluated between 2 and 8 h after SAH. Serum levels of TNFα, IL-1β, and IL-6 were assessed at 4 and 8 h after SAH. Anesthesia recovery time of all selected parameters was associated with severity of SAH. The consciousness recovery time test had the best predictive value, while the spontaneous ventilation recovery time test was able to bring information in the shortest time. The mRNA expressions of pro-inflammatory cytokines were significantly increased in severe SAH groups in both adjacent and distant areas. The inflammatory response in mild/moderate SAH groups was less strong, peaking at 4 h after SAH. Serum levels of pro-inflammatory cytokines were ambiguous. Anesthesia recovery time may be useful for bleeding severity prediction in the SAH model; however, further validation is needed. Severe subarachnoid hemorrhage is associated with the strong early inflammatory response, which has a global character, while mild subarachnoid hemorrhage is accompanied by a weaker inflammation.
Collapse
Affiliation(s)
- K Duris
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Department of Neurosurgery, The University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - J Lipkova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Z Splichal
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - T Madaraszova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Department of Neurosurgery, The University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Michal Jurajda
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| |
Collapse
|
25
|
Ran Y, Liu Z, Huang S, Shen J, Li F, Zhang W, Chen C, Geng X, Ji Z, Du H, Hu X. Splenectomy Fails to Provide Long-Term Protection Against Ischemic Stroke. Aging Dis 2018; 9:467-479. [PMID: 29896434 PMCID: PMC5988601 DOI: 10.14336/ad.2018.0130] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022] Open
Abstract
Splenectomy before or immediately after stroke provides early brain protection. This study aims to explore the effect of splenectomy on long-term neurological recovery after stroke, which is currently lacking in the field. Adult male rats were randomized into splenectomy or sham groups and then subjected to 90 min of middle cerebral artery occlusion (MCAO). Spleen was removed right upon reperfusion or 3d after MCAO. Infarct volume, neurological functions, and peripheral immune cell populations were assessed up to 28d after stroke. The results show that delayed removal of spleen did not reduce brain tissue loss and showed no effect on sensorimotor function (Rotarod, beam balance, forelimb placing, grid walk, and adhesive removal tests) or cognitive function (Morris water maze). Spleen removal immediately upon reperfusion, although significantly reduced the infarct size and immune cell infiltration 3d after MCAO, also failed to promote long-term recovery. Flow cytometry analysis demonstrated that immediate splenectomy after MCAO resulted in a prolonged decrease in the percentage of CD3+CD4+ and CD3+CD8+ T cells in total lymphocytes as compared to non-splenectomy MCAO rats. In contrast, the percentage of CD3-CD45RA+ B cells was significantly elevated after splenectomy. As a result, the ratio of T/B cells was significantly reduced in stroke rats with splenectomy. In conclusion, delayed splenectomy failed to provide long-term protection to the ischemic brain or improve functional recovery. The acute neuroprotective effect achieved by early splenectomy after stroke cannot last for long term. This loss of neuroprotection might be related to the prolonged disturbance in the T cell to B cell ratio.
Collapse
Affiliation(s)
- Yuanyuan Ran
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,2Central Laboratory, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zongjian Liu
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,2Central Laboratory, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Shuo Huang
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,2Central Laboratory, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Jiamei Shen
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Fengwu Li
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Wenxiu Zhang
- 2Central Laboratory, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Chen Chen
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zhili Ji
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Huishan Du
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaoming Hu
- 1China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China.,3Pittsburgh Institute of Brain Disorders and Recovery, and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| |
Collapse
|
26
|
Zhou Z, Lu J, Liu WW, Manaenko A, Hou X, Mei Q, Huang JL, Tang J, Zhang JH, Yao H, Hu Q. Advances in stroke pharmacology. Pharmacol Ther 2018; 191:23-42. [PMID: 29807056 DOI: 10.1016/j.pharmthera.2018.05.012] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Stroke occurs when a cerebral blood vessel is blocked or ruptured, and it is the major cause of death and adult disability worldwide. Various pharmacological agents have been developed for the treatment of stroke either through interrupting the molecular pathways leading to neuronal death or enhancing neuronal survival and regeneration. Except for rtPA, few of these agents have succeeded in clinical trials. Recently, with the understanding of the pathophysiological process of stroke, there is a resurrection of research on developing neuroprotective agents for stroke treatment, and novel molecular targets for neuroprotection and neurorestoration have been discovered to predict or offer clinical benefits. Here we review the latest major progress of pharmacological studies in stroke, especially in ischemic stroke; summarize emerging potential therapeutic mechanisms; and highlight recent clinical trials. The aim of this review is to provide a panorama of pharmacological interventions for stroke and bridge basic and translational research to guide the clinical management of stroke therapy.
Collapse
Affiliation(s)
- Zhenhua Zhou
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA; Department of Neurology, Southwest Hospital, the Third Military Medical University, Chongqing 400038, China
| | - Jianfei Lu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen-Wu Liu
- Department of Diving and Hyperbaric Medicine, the Second Military Medical University, Shanghai 200433, China
| | - Anatol Manaenko
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Xianhua Hou
- Department of Neurology, Southwest Hospital, the Third Military Medical University, Chongqing 400038, China
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, the Second Military Medical University, Shanghai 200003, China
| | - Jun-Long Huang
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China.
| | - Qin Hu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
27
|
Li B, Concepcion K, Meng X, Zhang L. Brain-immune interactions in perinatal hypoxic-ischemic brain injury. Prog Neurobiol 2017; 159:50-68. [PMID: 29111451 PMCID: PMC5831511 DOI: 10.1016/j.pneurobio.2017.10.006] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/26/2017] [Indexed: 01/07/2023]
Abstract
Perinatal hypoxia-ischemia remains the primary cause of acute neonatal brain injury, leading to a high mortality rate and long-term neurological deficits, such as behavioral, social, attentional, cognitive and functional motor deficits. An ever-increasing body of evidence shows that the immune response to acute cerebral hypoxia-ischemia is a major contributor to the pathophysiology of neonatal brain injury. Hypoxia-ischemia provokes an intravascular inflammatory cascade that is further augmented by the activation of resident immune cells and the cerebral infiltration of peripheral immune cells response to cellular damages in the brain parenchyma. This prolonged and/or inappropriate neuroinflammation leads to secondary brain tissue injury. Yet, the long-term effects of immune activation, especially the adaptive immune response, on the hypoxic-ischemic brain still remain unclear. The focus of this review is to summarize recent advances in the understanding of post-hypoxic-ischemic neuroinflammation triggered by the innate and adaptive immune responses and to discuss how these mechanisms modulate the brain vulnerability to injury. A greater understanding of the reciprocal interactions between the hypoxic-ischemic brain and the immune system will open new avenues for potential immunomodulatory therapy in the treatment of neonatal brain injury.
Collapse
Affiliation(s)
- Bo Li
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | - Katherine Concepcion
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Xianmei Meng
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
28
|
Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF, Shi Y. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol 2017; 163-164:144-171. [PMID: 28987927 DOI: 10.1016/j.pneurobio.2017.10.001] [Citation(s) in RCA: 643] [Impact Index Per Article: 80.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/30/2017] [Accepted: 10/02/2017] [Indexed: 01/06/2023]
Abstract
The blood-brain barrier (BBB) plays a vital role in regulating the trafficking of fluid, solutes and cells at the blood-brain interface and maintaining the homeostatic microenvironment of the CNS. Under pathological conditions, such as ischemic stroke, the BBB can be disrupted, followed by the extravasation of blood components into the brain and compromise of normal neuronal function. This article reviews recent advances in our knowledge of the mechanisms underlying BBB dysfunction and recovery after ischemic stroke. CNS cells in the neurovascular unit, as well as blood-borne peripheral cells constantly modulate the BBB and influence its breakdown and repair after ischemic stroke. The involvement of stroke risk factors and comorbid conditions further complicate the pathogenesis of neurovascular injury by predisposing the BBB to anatomical and functional changes that can exacerbate BBB dysfunction. Emphasis is also given to the process of long-term structural and functional restoration of the BBB after ischemic injury. With the development of novel research tools, future research on the BBB is likely to reveal promising potential therapeutic targets for protecting the BBB and improving patient outcome after ischemic stroke.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | | | - Ling Zhu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michael V L Bennett
- State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China; Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
29
|
Seifert HA, Benedek G, Liang J, Nguyen H, Kent G, Vandenbark AA, Saugstad JA, Offner H. Sex differences in regulatory cells in experimental stroke. Cell Immunol 2017; 318:49-54. [PMID: 28606360 DOI: 10.1016/j.cellimm.2017.06.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 06/06/2017] [Indexed: 12/21/2022]
Abstract
Stroke is the leading cause of disability in the United States. Sex differences, including smaller infarcts in females and greater involvement of immune-mediated inflammation in males may affect the efficacy of immune-modulating interventions. To address these differences, we sought to identify distinct stroke-modifying mechanisms in female vs. male mice. The current study demonstrated smaller infarcts and increased levels of regulatory CD19+CD5+CD1dhi B10 cells as well as anti-inflammatory CD11b+CD206+ microglia/macrophages in the ipsilateral vs. contralateral hemisphere of female but not male mice undergoing 60min middle cerebral artery occlusion followed by 96h of reperfusion. Moreover, female mice with MCAO had increased total spleen cell numbers but lower B10 levels in spleens. These results elucidate differing sex-dependent regulatory mechanisms that account for diminished stroke severity in females and underscore the need to test immune-modulating therapies for stroke in both males and females.
Collapse
Affiliation(s)
- Hilary A Seifert
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Gil Benedek
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Jian Liang
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ha Nguyen
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Gail Kent
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Arthur A Vandenbark
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA; Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Julie A Saugstad
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA; Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
30
|
Schuhmann MK, Langhauser F, Kraft P, Kleinschnitz C. B cells do not have a major pathophysiologic role in acute ischemic stroke in mice. J Neuroinflammation 2017; 14:112. [PMID: 28576128 PMCID: PMC5457733 DOI: 10.1186/s12974-017-0890-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/26/2017] [Indexed: 12/31/2022] Open
Abstract
Background Lymphocytes have been shown to play an important role in the pathophysiology of acute ischemic stroke, but the properties of B cells remain controversial. The aim of this study was to unravel the role of B cells during acute cerebral ischemia using pharmacologic B cell depletion, B cell transgenic mice, and adoptive B cell transfer experiments. Methods Transient middle cerebral artery occlusion (60 min) was induced in wild-type mice treated with an anti-CD20 antibody 24 h before stroke onset, JHD−/− mice and Rag1−/− mice 24 h after adoptive B cell transfer. Stroke outcome was assessed at days 1 and 3. Infarct volumes were calculated from 2,3,5-triphenyltetrazolium chloride (TTC)-stained brain sections, and neurological scores were evaluated. The local inflammatory response was determined by real-time PCR and immunohistochemistry. Apoptosis was analyzed by TUNEL staining, and astrocyte activation was revealed using immunohistochemistry and Western blot. Results Pharmacologic depletion of B cells did not influence infarct volumes and functional outcome at day 1 after stroke. Additionally, lack of circulating B cells in JHD−/− mice also failed to influence stroke outcome at days 1 and 3. Furthermore, reconstitution of Rag1−/− mice with B cells had no influence on infarct volumes. Conclusion Targeting B cells in experimental stroke did not influence lesion volume and functional outcome during the acute phase. Our findings argue against a major pathophysiologic role of B cells during acute ischemic stroke. Electronic supplementary material The online version of this article (doi:10.1186/s12974-017-0890-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Peter Kraft
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany. .,Department of Neurology, University Hospital Essen, 45147, Essen, Germany.
| |
Collapse
|
31
|
Santos Samary C, Pelosi P, Leme Silva P, Rieken Macedo Rocco P. Immunomodulation after ischemic stroke: potential mechanisms and implications for therapy. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2016; 20:391. [PMID: 27923376 PMCID: PMC5141640 DOI: 10.1186/s13054-016-1573-1] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Brain injuries are often associated with intensive care admissions, and carry high morbidity and mortality rates. Ischemic stroke is one of the most frequent causes of injury to the central nervous system. It is now increasingly clear that human stroke causes multi-organ systemic disease. Brain inflammation may lead to opposing local and systemic effects. Suppression of systemic immunity by the nervous system could protect the brain from additional inflammatory damage; however, it may increase the susceptibility to infection. Pneumonia and urinary tract infection are the most common complications occurring in patients after stroke. The mechanisms involved in lung-brain interactions are still unknown, but some studies have suggested that inhibition of the cholinergic anti-inflammatory pathway and release of glucocorticoids, catecholamines, and damage-associated molecular patterns (DAMPs) are among the pathophysiological mechanisms involved in communication from the ischemic brain to the lungs after stroke. This review describes the modifications in local and systemic immunity that occur after stroke, outlines mechanisms of stroke-induced immunosuppression and their role in pneumonia, and highlights potential therapeutic targets to reduce post-stroke complications. Despite significant advances towards a better understanding of the pathophysiology of ischemic stroke-induced immunosuppression and stroke-associated pneumonia (SAP) in recent years, many unanswered questions remain. The true incidence and outcomes of SAP, especially in intensive care unit settings, have yet to be determined, as has the full extent of stroke-induced immunosuppression and its clinical implications.
Collapse
Affiliation(s)
- Cynthia Santos Samary
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, 21941-902, Rio de Janeiro, RJ, Brazil
| | - Paolo Pelosi
- Department of Surgical Sciences and Integrated Diagnostics, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, 21941-902, Rio de Janeiro, RJ, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, 21941-902, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
32
|
Protection of Brain Injury by Amniotic Mesenchymal Stromal Cell-Secreted Metabolites. Crit Care Med 2016; 44:e1118-e1131. [DOI: 10.1097/ccm.0000000000001864] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
33
|
Kishimoto W, Nishikori M, Arima H, Miyoshi H, Sasaki Y, Kitawaki T, Shirakawa K, Kato T, Imaizumi Y, Ishikawa T, Ohno H, Haga H, Ohshima K, Takaori-Kondo A. Expression of Tim-1 in primary CNS lymphoma. Cancer Med 2016; 5:3235-3245. [PMID: 27709813 PMCID: PMC5119979 DOI: 10.1002/cam4.930] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 11/16/2022] Open
Abstract
Primary central nervous system lymphoma (PCNSL) is a distinct subtype of extranodal lymphoma with aggressive clinical course and poor outcome. As increased IL‐10/IL‐6 ratio is recognized in the cerebrospinal fluid (CSF) of PCNSL patients, we hypothesized that PCNSL might originate from a population of B cells with high IL‐10‐producing capacity, an equivalent of “regulatory B cells” in mice. We intended in this study to clarify whether Tim‐1, a molecule known as a marker for regulatory B cells in mice, is expressed in PCNSL. By immunohistochemical analysis, Tim‐1 was shown to be positive in as high as 54.2% of PCNSL (26 of 58 samples), while it was positive in 19.1% of systemic diffuse large B‐cell lymphoma (DLBCL) samples (17 of 89 samples; P < 0.001). Tim‐1 expression positively correlated with IL‐10 expression in PCNSL (Cramer's V = 0.55, P < 0.001), and forced expression of Tim‐1 in a PCNSL cell line resulted in increased IL‐10 secretion, suggesting that Tim‐1 is functionally linked with IL‐10 production in PCNSL. Moreover, soluble Tim‐1 was detectable in the CSF of PCNSL patients, and was suggested to parallel disease activity. In summary, PCNSL is characterized by frequent Tim‐1 expression, and its soluble form in CSF may become a useful biomarker for PCNSL.
Collapse
Affiliation(s)
- Wataru Kishimoto
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Momoko Nishikori
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Arima
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Miyoshi
- Department of Pathology, Kurume University School of Medicine, Asahimachi, Kurume, Fukuoka, Japan
| | - Yuya Sasaki
- Department of Pathology, Kurume University School of Medicine, Asahimachi, Kurume, Fukuoka, Japan
| | - Toshio Kitawaki
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kotaro Shirakawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeharu Kato
- Department of Hematology, Nagasaki University Hospital, Sakamoto, Nagasaki, Japan
| | - Yoshitaka Imaizumi
- Department of Hematology, Nagasaki University Hospital, Sakamoto, Nagasaki, Japan
| | - Takayuki Ishikawa
- Department of Hematology, Kobe City Medical Center General Hospital, Kobe, Hyogo, Japan
| | - Hitoshi Ohno
- Department of Hematology, Tenri Hospital, Mishima-cho, Tenri, Nara, Japan
| | - Hironori Haga
- Department of Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Asahimachi, Kurume, Fukuoka, Japan
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
34
|
Abstract
The immune response to acute cerebral ischemia is a major factor in stroke pathobiology and outcome. While the immune response starts locally in occluded and hypoperfused vessels and the ischemic brain parenchyma, inflammatory mediators generated in situ propagate through the organism as a whole. This "spillover" leads to a systemic inflammatory response first, followed by immunosuppression aimed at dampening the potentially harmful proinflammatory milieu. In this overview we will outline the inflammatory cascade from its starting point in the vasculature of the ischemic brain to the systemic immune response elicited by brain ischemia. Potential immunomodulatory therapeutic approaches, including preconditioning and immune cell therapy will also be discussed.
Collapse
Affiliation(s)
- Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
35
|
Selvaraj UM, Poinsatte K, Torres V, Ortega SB, Stowe AM. Heterogeneity of B Cell Functions in Stroke-Related Risk, Prevention, Injury, and Repair. Neurotherapeutics 2016; 13:729-747. [PMID: 27492770 PMCID: PMC5081124 DOI: 10.1007/s13311-016-0460-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is well established that post-stroke inflammation contributes to neurovascular injury, blood-brain barrier disruption, and poor functional recovery in both animal and clinical studies. However, recent studies also suggest that several leukocyte subsets, activated during the post-stroke immune response, can exhibit both pro-injury and pro-recovery phenotypes. In accordance with these findings, B lymphocytes, or B cells, play a heterogeneous role in the adaptive immune response to stroke. This review highlights what is currently understood about the various roles of B cells, with an emphasis on stroke risk factors, as well as post-stroke injury and repair. This includes an overview of B cell functions, such as antibody production, cytokine secretion, and contribution to the immune response as antigen presenting cells. Next, evidence for B cell-mediated mechanisms in stroke-related risk factors, including hypertension, diabetes, and atherosclerosis, is outlined, followed by studies that focus on B cells during endogenous protection from stroke. Subsequently, animal studies that investigate the role of B cells in post-stroke injury and repair are summarized, and the final section describes current B cell-related clinical trials for stroke, as well as other central nervous system diseases. This review reveals the complex role of B cells in stroke, with a focus on areas for potential clinical intervention for a disease that affects millions of people globally each year.
Collapse
Affiliation(s)
- Uma Maheswari Selvaraj
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Katherine Poinsatte
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Vanessa Torres
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Sterling B Ortega
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA
| | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, 6000 Harry Hines Blvd, MC8813, Dallas, TX, 75390, USA.
| |
Collapse
|
36
|
Regulatory T Cell Therapy for Ischemic Stroke: how far from Clinical Translation? Transl Stroke Res 2016; 7:415-9. [PMID: 27307291 DOI: 10.1007/s12975-016-0476-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 06/10/2016] [Indexed: 10/21/2022]
|
37
|
Shim R, Wong CHY. Ischemia, Immunosuppression and Infection--Tackling the Predicaments of Post-Stroke Complications. Int J Mol Sci 2016; 17:ijms17010064. [PMID: 26742037 PMCID: PMC4730309 DOI: 10.3390/ijms17010064] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/14/2015] [Accepted: 12/24/2015] [Indexed: 12/29/2022] Open
Abstract
The incidence of stroke has risen over the past decade and will continue to be one of the leading causes of death and disability worldwide. While a large portion of immediate death following stroke is due to cerebral infarction and neurological complications, the most common medical complication in stroke patients is infection. In fact, infections, such as pneumonia and urinary tract infections, greatly worsen the clinical outcome of stroke patients. Recent evidence suggests that the disrupted interplay between the central nervous system and immune system contributes to the development of infection after stroke. The suppression of systemic immunity by the nervous system is thought to protect the brain from further inflammatory insult, yet this comes at the cost of increased susceptibility to infection after stroke. To improve patient outcome, there have been attempts to lessen the stroke-associated bacterial burden through the prophylactic use of broad-spectrum antibiotics. However, preventative antibiotic treatments have been unsuccessful, and therefore have been discouraged. Additionally, with the ever-rising obstacle of antibiotic-resistance, future therapeutic options to reverse immune impairment after stroke by augmentation of host immunity may be a viable alternative option. However, cautionary steps are required to ensure that collateral ischemic damage caused by cerebral inflammation remains minimal.
Collapse
Affiliation(s)
- Raymond Shim
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, VIC 3168, Australia.
| | - Connie H Y Wong
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, Monash University, Clayton, VIC 3168, Australia.
| |
Collapse
|
38
|
Anrather J, Iadecola C, Hallenbeck J. Inflammation and Immune Response. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
39
|
Adenosine A1 receptors contribute to immune regulation after neonatal hypoxic ischemic brain injury. Purinergic Signal 2015; 12:89-101. [PMID: 26608888 DOI: 10.1007/s11302-015-9482-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/28/2015] [Indexed: 12/20/2022] Open
Abstract
Neonatal brain hypoxic ischemia (HI) often results in long-term motor and cognitive impairments. Post-ischemic inflammation greatly effects outcome and adenosine receptor signaling modulates both HI and immune cell function. Here, we investigated the influence of adenosine A1 receptor deficiency (A1R(-/-)) on key immune cell populations in a neonatal brain HI model. Ten-day-old mice were subjected to HI. Functional outcome was assessed by open locomotion and beam walking test and infarction size evaluated. Flow cytometry was performed on brain-infiltrating cells, and semi-automated analysis of flow cytometric data was applied. A1R(-/-) mice displayed larger infarctions (+33%, p < 0.05) and performed worse in beam walking tests (44% more mistakes, p < 0.05) than wild-type (WT) mice. Myeloid cell activation after injury was enhanced in A1R(-/-) versus WT brains. Activated B lymphocytes expressing IL-10 infiltrated the brain after HI in WT, but were less activated and did not increase in relative frequency in A1R(-/-). Also, A1R(-/-) B lymphocytes expressed less IL-10 than their WT counterparts, the A1R antagonist DPCPX decreased IL-10 expression whereas the A1R agonist CPA increased it. CD4(+) T lymphocytes including FoxP3(+) T regulatory cells, were unaffected by genotype, whereas CD8(+) T lymphocyte responses were smaller in A1R(-/-) mice. Using PCA to characterize the immune profile, we could discriminate the A1R(-/-) and WT genotypes as well as sham operated from HI-subjected animals. We conclude that A1R signaling modulates IL-10 expression by immune cells, influences the activation of these cells in vivo, and affects outcome after HI.
Collapse
|
40
|
Bodhankar S, Chen Y, Lapato A, Vandenbark AA, Murphy SJ, Saugstad JA, Offner H. Regulatory CD8(+)CD122 (+) T-cells predominate in CNS after treatment of experimental stroke in male mice with IL-10-secreting B-cells. Metab Brain Dis 2015; 30:911-924. [PMID: 25537181 PMCID: PMC4481189 DOI: 10.1007/s11011-014-9639-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/02/2014] [Indexed: 12/31/2022]
Abstract
Clinical stroke induces inflammatory processes leading to cerebral and splenic injury and profound peripheral immunosuppression. IL-10 expression is elevated during major CNS diseases and limits inflammation in the brain. Recent evidence demonstrated that transfer of IL-10(+) B-cells reduced infarct volume in male C57BL/6J (wild-type, WT) recipient mice when given 24 h prior to or 4 h after middle cerebral artery occlusion (MCAO). The purpose of this study was to determine if passively transferred IL-10(+) B-cells can exert therapeutic and immunoregulatory effects when injected 24 h after MCAO induction in B-cell-sufficient male WT mice. The results demonstrated that IL-10(+) B-cell treated mice had significantly reduced infarct volumes in the ipsilateral cortex and hemisphere and improved neurological deficits vs. Vehicle-treated control mice after 60 min occlusion and 96 h of reperfusion. The MCAO-protected B-cell recipient mice had less splenic atrophy and reduced numbers of activated, inflammatory T-cells, decreased infiltration of T-cells and a less inflammatory milieu in the ischemic hemispheres compared with Vehicle-treated control mice. These immunoregulatory changes occurred in concert with the predominant appearance of IL-10-secreting CD8(+)CD122(+) Treg cells in both the spleen and the MCAO-affected brain hemisphere. This study for the first time demonstrates a major neuroprotective role for IL-10(+) B-cells in treating MCAO in male WT mice at a time point well beyond the ~4 h tPA treatment window, leading to the generation of a dominant IL-10(+)CD8(+)CD122(+) Treg population associated with spleen preservation and reduced CNS inflammation.
Collapse
Affiliation(s)
- Sheetal Bodhankar
- Neuroimmunology Research, VA Medical Center, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Yingxin Chen
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Andrew Lapato
- Neuroimmunology Research, VA Medical Center, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Arthur A. Vandenbark
- Neuroimmunology Research, VA Medical Center, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Stephanie J. Murphy
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Neuroimmunology Research, VA Medical Center, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
41
|
Ma S, Zhao H, Ji X, Luo Y. Peripheral to central: Organ interactions in stroke pathophysiology. Exp Neurol 2015; 272:41-9. [PMID: 26054885 DOI: 10.1016/j.expneurol.2015.05.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 05/20/2015] [Accepted: 05/23/2015] [Indexed: 12/27/2022]
Abstract
Stroke is associated with a high risk of disability and mortality, and with the exception of recombinant tissue-type plasminogen activator for acute stroke, most treatments have proven ineffective. Clinical translation of promising experimental therapeutics is limited by inadequate stroke models and a lack of understanding of the mechanisms underlying acute stroke and how they affect outcome. Bidirectional communication between the ischemic brain and peripheral immune system modulates stroke progression and tissue repair, while epidemiological studies have provided evidence of an association between organ dysfunction and stroke risk. This crosstalk can determine the fate of stroke patients and must be taken into consideration when investigating the pathophysiological mechanisms and therapeutic options for stroke. This review summarizes the current evidence for interactions between the brain and other organs in stroke pathophysiology in basic and clinic studies, and discusses the role of these interactions in the progression and outcome of stroke and how they can direct the development of more effective treatment strategies.
Collapse
Affiliation(s)
- Shubei Ma
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Haiping Zhao
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing 100053, China.
| | - Yumin Luo
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Center of Stroke, Beijing Institute for Brain Disorders, Beijing 100053, China.
| |
Collapse
|
42
|
Liesz A, Hu X, Kleinschnitz C, Offner H. Functional role of regulatory lymphocytes in stroke: facts and controversies. Stroke 2015; 46:1422-30. [PMID: 25791715 DOI: 10.1161/strokeaha.114.008608] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 02/24/2015] [Indexed: 01/02/2023]
Affiliation(s)
- Arthur Liesz
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (A.L.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (A.L.); Department of Neurology, University of Pittsburgh, PA (X.H.); Department of Neurology, University Hospital Würzburg, Würzburg, Germany (C.K.); Department of Neurology and Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland (H.O.); and Neuroimmunology Research, Portland, OR (H.O.).
| | - Xiaoming Hu
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (A.L.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (A.L.); Department of Neurology, University of Pittsburgh, PA (X.H.); Department of Neurology, University Hospital Würzburg, Würzburg, Germany (C.K.); Department of Neurology and Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland (H.O.); and Neuroimmunology Research, Portland, OR (H.O.)
| | - Christoph Kleinschnitz
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (A.L.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (A.L.); Department of Neurology, University of Pittsburgh, PA (X.H.); Department of Neurology, University Hospital Würzburg, Würzburg, Germany (C.K.); Department of Neurology and Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland (H.O.); and Neuroimmunology Research, Portland, OR (H.O.)
| | - Halina Offner
- From the Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany (A.L.); Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (A.L.); Department of Neurology, University of Pittsburgh, PA (X.H.); Department of Neurology, University Hospital Würzburg, Würzburg, Germany (C.K.); Department of Neurology and Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland (H.O.); and Neuroimmunology Research, Portland, OR (H.O.)
| |
Collapse
|
43
|
Abstract
Innovation is a form of purposeful discovery behavior that exploits the unexpected, utilizes imagination, and provides one avenue of new solutions to complex human health needs. It is through this lens that two examples are described in which innovative approaches have been used to dissect the complexities of stroke pathophysiology. The first example focuses on one of the most fundamental genetic factors relevant to the brain and ischemic injury: biological sex. Much might be gained by understanding the details of sex-specific pathobiology, if the field is to develop therapies that work well in patients of both sexes. The second example surrounds brain-spleen cell cycling after stroke which is fundamental to our evolving understanding that stroke is a systemic disease, rather than solely a lesion of the brain. While much work remains, it is now apparent that brain-spleen cell cycling is temporally specific, varies in intensity, and involves cell players that are of much wider lineages than originally believed. In the future, it is likely that innovation will need to turn to “big data”, particularly if our field is to tackle the daunting questions that most greatly matter to unraveling brain injury. The huge availability and growth rate of biomedical data, handled in a shared but coherent environment, offers an opportunity to further vitalize stroke research.
Collapse
|
44
|
The authors reply. Crit Care Med 2014; 42:e804-5. [PMID: 25402311 DOI: 10.1097/ccm.0000000000000652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Targeting the peripheral inflammatory response to stroke: role of the spleen. Transl Stroke Res 2014; 5:635-7. [PMID: 25252625 DOI: 10.1007/s12975-014-0372-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 09/15/2014] [Indexed: 12/12/2022]
|
46
|
Ries S, Hilgenberg E, Lampropoulou V, Shen P, Dang VD, Wilantri S, Sakwa I, Fillatreau S. B-type suppression: a role played by "regulatory B cells" or "regulatory plasma cells"? Eur J Immunol 2014; 44:1251-7. [PMID: 24615065 DOI: 10.1002/eji.201343683] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 01/17/2014] [Accepted: 03/06/2014] [Indexed: 01/10/2023]
Abstract
B-cell depletion can improve disease in some patients with rheumatoid arthritis or multiple sclerosis, indicating the pathogenic contribution of B cells to autoimmunity. However, studies in mice have demonstrated that B cells have immunosuppressive functions as well, with IL-10 being a critical mediator of B-cell-mediated suppression. IL-10-secreting B cells have been shown to promote disease remission in some mouse models of autoimmune disorders. Human B cells also produce IL-10, and evidence is accumulating that human IL-10-producing B cells might inhibit immunity. There is considerable interest in identifying the phenotype of B cells providing IL-10 in a suppressive manner, which would facilitate the analysis of the molecular mechanisms controlling this B-cell property. Here, we review current knowledge on the B-cell subpopulations found to provide suppressive functions in mice, considering both the pathological context in which they were identified and the signals that control their induction. We discuss the phenotype of B cells that have IL-10-dependent regulatory activities in mice, which leads us to propose that antibody-secreting cells are, in some cases at least, the major source of B-cell-derived regulatory IL-10 in vivo. Anti-inflammatory cytokine production by antibody-secreting cells offers a novel mechanism for the coordination of innate and humoral immune responses.
Collapse
Affiliation(s)
- Stefanie Ries
- Deutsches Rheuma-Forschungszentrum, a Leibniz Institute Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Chu HX, Kim HA, Lee S, Moore JP, Chan CT, Vinh A, Gelderblom M, Arumugam TV, Broughton BRS, Drummond GR, Sobey CG. Immune cell infiltration in malignant middle cerebral artery infarction: comparison with transient cerebral ischemia. J Cereb Blood Flow Metab 2014; 34:450-9. [PMID: 24326388 PMCID: PMC3948121 DOI: 10.1038/jcbfm.2013.217] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 11/04/2013] [Accepted: 11/05/2013] [Indexed: 12/29/2022]
Abstract
We tested whether significant leukocyte infiltration occurs in a mouse model of permanent cerebral ischemia. C57BL6/J male mice underwent either permanent (3 or 24 hours) or transient (1 or 2 hours+22- to 23-hour reperfusion) middle cerebral artery occlusion (MCAO). Using flow cytometry, we observed ∼15,000 leukocytes (CD45(+high) cells) in the ischemic hemisphere as early as 3 hours after permanent MCAO (pMCAO), comprising ∼40% lymphoid cells and ∼60% myeloid cells. Neutrophils were the predominant cell type entering the brain, and were increased to ∼5,000 as early as 3 hours after pMCAO. Several cell types (monocytes, macrophages, B lymphocytes, CD8(+) T lymphocytes, and natural killer cells) were also increased at 3 hours to levels sustained for 24 hours, whereas others (CD4(+) T cells, natural killer T cells, and dendritic cells) were unchanged at 3 hours, but were increased by 24 hours after pMCAO. Immunohistochemical analysis revealed that leukocytes typically had entered and widely dispersed throughout the parenchyma of the infarct within 3 hours. Moreover, compared with pMCAO, there were ∼50% fewer infiltrating leukocytes at 24 hours after transient MCAO (tMCAO), independent of infarct size. Microglial cell numbers were bilaterally increased in both models. These findings indicate that a profound infiltration of inflammatory cells occurs in the brain early after focal ischemia, especially without reperfusion.
Collapse
Affiliation(s)
- Hannah X Chu
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Hyun Ah Kim
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Seyoung Lee
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Jeffrey P Moore
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Christopher T Chan
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Antony Vinh
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thiruma V Arumugam
- Department of Pharmacology, University of Queensland, St Lucia, Queensland, Australia
| | - Brad R S Broughton
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Grant R Drummond
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Christopher G Sobey
- Vascular Biology and Immunopharmacology Group, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
48
|
Bodhankar S, Chen Y, Vandenbark AA, Murphy SJ, Offner H. Treatment of experimental stroke with IL-10-producing B-cells reduces infarct size and peripheral and CNS inflammation in wild-type B-cell-sufficient mice. Metab Brain Dis 2014; 29:59-73. [PMID: 24374817 PMCID: PMC3944055 DOI: 10.1007/s11011-013-9474-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 12/16/2013] [Indexed: 12/11/2022]
Abstract
Clinical stroke induces inflammatory processes leading to cerebral and splenic injury and profound peripheral immunosuppression. IL-10 expression is elevated during major CNS diseases and limits inflammation in the brain. Recent evidence demonstrated that absence of B-cells led to larger infarct volumes and CNS damage after middle cerebral artery occlusion (MCAO) that could be prevented by transfer of IL-10(+) B-cells. The purpose of this study was to determine if the beneficial immunoregulatory effects on MCAO of the IL-10(+) B-cell subpopulation also extends to B-cell-sufficient mice that would better represent stroke subjects. CNS inflammation and infarct volumes were evaluated in male C57BL/6J (WT) mice that received either RPMI or IL-10(+) B-cells and underwent 60 min of middle cerebral artery occlusion (MCAO) followed by 96 h of reperfusion. Transfer of IL-10(+) B-cells markedly reduced infarct volume in WT recipient mice when given 24 h prior to or 4 h after MCAO. B-cell protected (24 h pre-MCAO) mice had increased regulatory subpopulations in the periphery, reduced numbers of activated, inflammatory T-cells, decreased infiltration of T-cells and a less inflammatory milieu in the ischemic hemispheres of the IL-10(+) B-cell-treated group. Moreover, transfer of IL-10(+) B-cells 24 h before MCAO led to a significant preservation of regulatory immune subsets in the IL-10(+) B-cell protected group presumably indicating their role in immunomodulatory mechanisms, post-stroke. Our studies are the first to demonstrate a major immunoregulatory role for IL-10(+) regulatory B-cells in preventing and treating MCAO in WT mice and also implicating their potential role in attenuating complications due to post-stroke immunosuppression.
Collapse
Affiliation(s)
- Sheetal Bodhankar
- Neuroimmunology Research, VA Medical Center, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Yingxin Chen
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Arthur A. Vandenbark
- Neuroimmunology Research, VA Medical Center, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Stephanie J. Murphy
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Halina Offner
- Neuroimmunology Research, VA Medical Center, Portland, OR, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
49
|
Monson NL, Ortega SB, Ireland SJ, Meeuwissen AJ, Chen D, Plautz EJ, Shubel E, Kong X, Li MK, Freriks LH, Stowe AM. Repetitive hypoxic preconditioning induces an immunosuppressed B cell phenotype during endogenous protection from stroke. J Neuroinflammation 2014; 11:22. [PMID: 24485041 PMCID: PMC3926678 DOI: 10.1186/1742-2094-11-22] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 01/13/2014] [Indexed: 12/16/2022] Open
Abstract
Background Repetitive hypoxic preconditioning (RHP) creates an anti-inflammatory phenotype that protects from stroke-induced injury for months after a 2-week treatment. The mechanisms underlying long-term tolerance are unknown, though one exposure to hypoxia significantly increased peripheral B cell representation. For this study, we sought to determine if RHP specifically recruited B cells into the protected ischemic hemisphere, and whether RHP could phenotypically alter B cells prior to stroke onset. Methods Adult, male SW/ND4 mice received RHP (nine exposures over 2 weeks; 8 to 11 % O2; 2 to 4 hours) or identical exposures to 21 % O2 as control. Two weeks following RHP, a 60-minute transient middle cerebral artery occlusion was induced. Standard techniques quantified CXCL13 mRNA and protein expression. Two days after stroke, leukocytes were isolated from brain tissue (70:30 discontinuous Percoll gradient) and profiled on a BD-FACS Aria flow cytometer. In a separate cohort without stroke, sorted splenic CD19+ B cells were isolated 2 weeks after RHP and analyzed on an Illumina MouseWG-6 V2 Bead Chip. Final gene pathways were determined using Ingenuity Pathway Analysis. Student’s t-test or one-way analysis of variance determined significance (P < 0.05). Results CXCL13, a B cell-specific chemokine, was upregulated in post-stroke cortical vessels of both groups. In the ischemic hemisphere, RHP increased B cell representation by attenuating the diapedesis of monocyte, macrophage, neutrophil and T cells, to quantities indistinguishable from the uninjured, contralateral hemisphere. Pre-stroke splenic B cells isolated from RHP-treated mice had >1,900 genes differentially expressed by microarray analysis. Genes related to B-T cell interactions, including antigen presentation, B cell differentiation and antibody production, were profoundly downregulated. Maturation and activation were arrested in a cohort of B cells from pre-stroke RHP-treated mice while regulatory B cells, a subset implicated in neurovascular protection from stroke, were upregulated. Conclusions Collectively, our data characterize an endogenous neuroprotective phenotype that utilizes adaptive immune mechanisms pre-stroke to protect the brain from injury post-stroke. Future studies to validate the role of B cells in minimizing injury and promoting central nervous system recovery, and to determine whether B cells mediate an adaptive immunity to systemic hypoxia that protects from subsequent stroke, are needed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Ann M Stowe
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center at Dallas, 6000 Harry Hines Blvd, Dallas, TX 75390-8813, USA.
| |
Collapse
|
50
|
Vogelgesang A, Becker KJ, Dressel A. Immunological consequences of ischemic stroke. Acta Neurol Scand 2014; 129:1-12. [PMID: 23848237 DOI: 10.1111/ane.12165] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2013] [Indexed: 12/24/2022]
Abstract
The treatment of ischemic stroke is one of the great challenges in modern neurology. The localization and the size of the infarct determine the long-term disability of stroke survivors. Recent observations have revealed that stroke also alters the function of the immune system and vice versa: At the site of the infarct, a local inflammatory response develops that enhances brain lesion development. In experimental stroke, proof-of-concept studies confirm that inhibition of this immune response reduces lesion volume and improves outcome. In the peripheral blood of stroke patients, though, lymphocytopenia and monocyte dysfunction develop. These changes reflect a clinically relevant impairment of bacterial defense mechanisms because they are associated with an enhanced risk to acquire post-stroke infections. Stress hormones have been identified as important mediators of stroke-induced immune suppression. The pharmacological inhibition of beta adrenergic receptors, but not the inhibition of steroids, is effective in reducing infection and improving clinical outcome in experimental stroke; catecholamine release therefore appears causally related to stroke-induced immune suppression. Strong evidence supports the hypothesis that these immune alterations impact the clinical course of stroke patients. Thus, the development of new therapeutic strategies targeted to alter the immunological consequences of stroke appears promising. However, to date, the beneficial effects seen in experimental stroke have not been successfully translated into a clinical trial. This brief review summarizes the current understanding of the immunological consequences of ischemic stroke. Finally, we propose a concept that links the peripheral immune suppression with the development of local inflammation.
Collapse
Affiliation(s)
- A. Vogelgesang
- Universitiy Medicine; Institute of Immunology and Transfusion Medicine; Greifswald Germany
| | - K. J. Becker
- University of Washington School of Medicine; Harborview Medical Center; Seattle WA USA
| | - A. Dressel
- Section of Neuroimmunology; Department of Neurology, University Medicine Greifswald; Greifswald Germany
| |
Collapse
|