51
|
Wang X, Chacon LI, Derakhshandeh R, Rodriguez HJ, Han DD, Kostyushev DS, Henry TD, Traverse JH, Moyé L, Simari RD, Taylor DA, Springer ML. Impaired therapeutic efficacy of bone marrow cells from post-myocardial infarction patients in the TIME and LateTIME clinical trials. PLoS One 2020; 15:e0237401. [PMID: 32841277 PMCID: PMC7446972 DOI: 10.1371/journal.pone.0237401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/25/2020] [Indexed: 01/07/2023] Open
Abstract
Implantation of bone marrow-derived cells (BMCs) into mouse hearts post-myocardial infarction (MI) limits cardiac functional decline. However, clinical trials of post-MI BMC therapy have yielded conflicting results. While most laboratory experiments use healthy BMC donor mice, clinical trials use post-MI autologous BMCs. Post-MI mouse BMCs are therapeutically impaired, due to inflammatory changes in BMC composition. Thus, therapeutic efficacy of the BMCs progressively worsens after MI but recovers as donor inflammatory response resolves. The availability of post-MI patient BM mononuclear cells (MNCs) from the TIME and LateTIME clinical trials enabled us to test if human post-MI MNCs undergo a similar period of impaired efficacy. We hypothesized that MNCs from TIME trial patients would be less therapeutic than healthy human donor MNCs when implanted into post-MI mouse hearts, and that therapeutic properties would be restored in MNCs from LateTIME trial patients. Post-MI SCID mice received MNCs from healthy donors, TIME patients, or LateTIME patients. Cardiac function improved considerably in the healthy donor group, but neither the TIME nor LateTIME group showed therapeutic effect. Conclusion: post-MI human MNCs lack therapeutic benefits possessed by healthy MNCs, which may partially explain why BMC clinical trials have been less successful than mouse studies.
Collapse
Affiliation(s)
- Xiaoyin Wang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States of America
| | | | - Ronak Derakhshandeh
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States of America
| | - Hilda J. Rodriguez
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States of America
| | - Daniel D. Han
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Dmitry S. Kostyushev
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, United States of America
| | - Timothy D. Henry
- The Carl and Edyth Lindner Center for Research and Education at The Christ Hospital, Cincinnati, OH, United States of America
| | - Jay H. Traverse
- Minneapolis Heart Institute Foundation, Minneapolis, MN, United States of America
| | - Lem Moyé
- University of Texas Health School of Public Health, Houston, TX, United States of America
| | - Robert D. Simari
- Kansas University Medical Center, Kansas City, KS, United States of America
| | - Doris A. Taylor
- Texas Heart Institute, Houston, TX, United States of America
| | - Matthew L. Springer
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States of America
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
52
|
Iadecola C, Buckwalter MS, Anrather J. Immune responses to stroke: mechanisms, modulation, and therapeutic potential. J Clin Invest 2020; 130:2777-2788. [PMID: 32391806 PMCID: PMC7260029 DOI: 10.1172/jci135530] [Citation(s) in RCA: 372] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Stroke is the second leading cause of death worldwide and a leading cause of disability. Most strokes are caused by occlusion of a major cerebral artery, and substantial advances have been made in elucidating how ischemia damages the brain. In particular, increasing evidence points to a double-edged role of the immune system in stroke pathophysiology. In the acute phase, innate immune cells invade brain and meninges and contribute to ischemic damage, but may also be protective. At the same time, danger signals released into the circulation by damaged brain cells lead to activation of systemic immunity, followed by profound immunodepression that promotes life-threatening infections. In the chronic phase, antigen presentation initiates an adaptive immune response targeted to the brain, which may underlie neuropsychiatric sequelae, a considerable cause of poststroke morbidity. Here, we briefly review these pathogenic processes and assess the potential therapeutic value of targeting immunity in human stroke.
Collapse
Affiliation(s)
- Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Marion S. Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University Medical Center, Stanford, California, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
53
|
Zera KA, Buckwalter MS. The Local and Peripheral Immune Responses to Stroke: Implications for Therapeutic Development. Neurotherapeutics 2020; 17:414-435. [PMID: 32193840 PMCID: PMC7283378 DOI: 10.1007/s13311-020-00844-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The immune response to stroke is an exciting target for future stroke therapies. Stroke is a leading cause of morbidity and mortality worldwide, and clot removal (mechanical or pharmacological) to achieve tissue reperfusion is the only therapy currently approved for patient use. Due to a short therapeutic window and incomplete effectiveness, however, many patients are left with infarcted tissue that stimulates inflammation. Although this is critical to promote repair, it can also damage surrounding healthy brain tissue. In addition, acute immunodepression and subsequent infections are common and are associated with worse patient outcomes. Thus, the acute immune response is a major focus of researchers attempting to identify ways to amplify its benefits and suppress its negative effects to improve short-term recovery of patients. Here we review what is known about this powerful process. This includes the role of brain resident cells such as microglia, peripherally activated cells such as macrophages and neutrophils, and activated endothelium. The role of systemic immune activation and subsequent immunodepression in the days after stroke is also discussed, as is the chronic immune responses and its effects on cognitive function. The biphasic role of inflammation, as well as complex timelines of cell production, differentiation, and trafficking, suggests that the relationship between the acute and chronic phases of stroke recovery is complex. Gaining a more complete understanding of this intricate process by which inflammation is initiated, propagated, and terminated may potentially lead to therapeutics that can treat a larger population of stroke patients than what is currently available. The immune response plays a critical role in patient recovery in both the acute and chronic phases after stroke. In patients, the immune response can be beneficial by promoting repair and recovery, and also detrimental by propagating a pro-inflammatory microenvironment. Thus, it is critical to understand the mechanisms of immune activation following stroke in order to successfully design therapeutics.
Collapse
Affiliation(s)
- Kristy A Zera
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Department of Neurosurgery, Stanford Univeristy School of Medicine, Stanford, CA, USA.
| |
Collapse
|
54
|
Sun J, Meng D, Liu Z, Hua X, Xu Z, Zhu J, Qian Z, Xu X. Neutrophil to Lymphocyte Ratio Is a Therapeutic Biomarker for Spontaneous Hemorrhagic Transformation. Neurotox Res 2020; 38:219-227. [PMID: 32157627 DOI: 10.1007/s12640-020-00181-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/31/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Jing Sun
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Danyang Meng
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Zhaoxia Liu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Xiaodong Hua
- The Medical College of Georgia at Augusta University, AU/UGA Medical Partnership, Athens, GA, 30602, USA
| | - Zhiqiang Xu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Jiangtao Zhu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China
| | - Zhiyuan Qian
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China.
| | - Xingshun Xu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu, China.
- Institute of Neuroscience, Soochow University, Suzhou City, 215123, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou City, 215123, Jiangsu, China.
| |
Collapse
|
55
|
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: 50] [Impact Index Per Article: 12.5] [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
|
56
|
B cells migrate into remote brain areas and support neurogenesis and functional recovery after focal stroke in mice. Proc Natl Acad Sci U S A 2020; 117:4983-4993. [PMID: 32051245 PMCID: PMC7060723 DOI: 10.1073/pnas.1913292117] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Neuroinflammation occurs immediately after stroke onset in the ischemic infarct, but whether neuroinflammation occurs in remote regions supporting plasticity and functional recovery remains unknown. We used advanced imaging to quantify whole-brain diapedesis of B cells, an immune cell capable of producing neurotrophins. We identify bilateral B cell diapedesis into remote regions, outside of the injury, that support motor and cognitive recovery in young male mice. Poststroke depletion of B cells confirms a positive role in neurogenesis, neuronal survival, and recovery of motor coordination, spatial learning, and anxiety. More than 80% of stroke survivors have long-term disability uniquely affected by age and lifestyle factors. Thus, identifying beneficial neuroinflammation during long-term recovery increases the opportunity of therapeutic interventions to support functional recovery. Lymphocytes infiltrate the stroke core and penumbra and often exacerbate cellular injury. B cells, however, are lymphocytes that do not contribute to acute pathology but can support recovery. B cell adoptive transfer to mice reduced infarct volumes 3 and 7 d after transient middle cerebral artery occlusion (tMCAo), independent of changing immune populations in recipient mice. Testing a direct neurotrophic effect, B cells cocultured with mixed cortical cells protected neurons and maintained dendritic arborization after oxygen-glucose deprivation. Whole-brain volumetric serial two-photon tomography (STPT) and a custom-developed image analysis pipeline visualized and quantified poststroke B cell diapedesis throughout the brain, including remote areas supporting functional recovery. Stroke induced significant bilateral B cell diapedesis into remote brain regions regulating motor and cognitive functions and neurogenesis (e.g., dentate gyrus, hypothalamus, olfactory areas, cerebellum) in the whole-brain datasets. To confirm a mechanistic role for B cells in functional recovery, rituximab was given to human CD20+ (hCD20+) transgenic mice to continuously deplete hCD20+-expressing B cells following tMCAo. These mice experienced delayed motor recovery, impaired spatial memory, and increased anxiety through 8 wk poststroke compared to wild type (WT) littermates also receiving rituximab. B cell depletion reduced stroke-induced hippocampal neurogenesis and cell survival. Thus, B cell diapedesis occurred in areas remote to the infarct that mediated motor and cognitive recovery. Understanding the role of B cells in neuronal health and disease-based plasticity is critical for developing effective immune-based therapies for protection against diseases that involve recruitment of peripheral immune cells into the injured brain.
Collapse
|
57
|
Chen KY, Wu KC, Hueng DY, Huang KF, Pang CY. Anti-inflammatory effects of powdered product of Bu Yang Huan Wu decoction: Possible role in protecting against Transient Focal Cerebral Ischemia. Int J Med Sci 2020; 17:1854-1863. [PMID: 32714088 PMCID: PMC7378667 DOI: 10.7150/ijms.46581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/17/2020] [Indexed: 12/25/2022] Open
Abstract
Bu Yang Huan Wu decoction (BYHW) is a traditional Chinese medicine (TCM) that consists of several herbs and has been used in patients with ischemic stroke for centuries. Although powdered formula of BYHW has widely been prescribed in clinic nowadays, evidence-based effectiveness and mechanism of action of BYHW powdered product in stroke remain to be characterized. Adult male Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 90 min followed by reperfusion for 24 h (ischemia/reperfusion; I/R) or sham surgery. After I/R, the rats were then given low dose (0.5 g/kg) and high dose (2.5 g/kg) of BYHW or vehicle by oral gavage twice a day for seven consecutive days. The results showed that I/R induced obvious cerebral infarction and neurobehavioral defects, in parallel with histological aberrations and extensive signaling of proinflammatory cytokines, including tumor necrosis factor (TNF-α) and interleukin-6 (IL-6), in the stroke model. Post-I/R treatment with BYHW powdered product significantly reduced the infarct area and ameliorated neurofunctional defects in a dose-dependent manner. The dose dependence was associated with TNF-α downregulation and interleukin-10 (IL-10) induction. In summary, the present findings demonstrated that BYHW powdered product exhibited therapeutic efficacy for experimental stroke and a higher dose treatment may strengthen the effectiveness via inflammatory modulation.
Collapse
Affiliation(s)
- Kuan-Yu Chen
- Department of Surgery, New Taipei City Hospital, New Taipei city, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien city, Taiwan
| | - Kuo-Chen Wu
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Dueng-Yuan Hueng
- Department of Neurosurgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Feng Huang
- School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan.,Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Medical Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Cheng-Yoong Pang
- Institute of Medical Sciences, Tzu Chi University, Hualien city, Taiwan.,Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien city, Taiwan
| |
Collapse
|
58
|
Activated CD8+ T Cells Cause Long-Term Neurological Impairment after Traumatic Brain Injury in Mice. Cell Rep 2019; 29:1178-1191.e6. [DOI: 10.1016/j.celrep.2019.09.046] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 08/08/2019] [Accepted: 09/16/2019] [Indexed: 12/28/2022] Open
|
59
|
Jian Z, Liu R, Zhu X, Smerin D, Zhong Y, Gu L, Fang W, Xiong X. The Involvement and Therapy Target of Immune Cells After Ischemic Stroke. Front Immunol 2019; 10:2167. [PMID: 31572378 PMCID: PMC6749156 DOI: 10.3389/fimmu.2019.02167] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/28/2019] [Indexed: 12/24/2022] Open
Abstract
After ischemic stroke, the integrity of the blood-brain barrier is compromised. Peripheral immune cells, including neutrophils, T cells, B cells, dendritic cells, and macrophages, infiltrate into the ischemic brain tissue and play an important role in regulating the progression of ischemic brain injury. In this review, we will discuss the role of different immune cells after stroke in the secondary inflammatory reaction and focus on the phenotypes and functions of macrophages in ischemic stroke, as well as briefly introduce the anti-ischemic stroke therapy targeting macrophages.
Collapse
Affiliation(s)
- Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Liu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Pharmacology and Toxicology, Shandong Institute for Food and Drug Control, Jinan, China
| | - Xiqun Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Daniel Smerin
- Department of Neurosurgery, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
60
|
Zhao J, Mu H, Liu L, Jiang X, Wu D, Shi Y, Leak RK, Ji X. Transient selective brain cooling confers neurovascular and functional protection from acute to chronic stages of ischemia/reperfusion brain injury. J Cereb Blood Flow Metab 2019; 39:1215-1231. [PMID: 30334662 PMCID: PMC6668511 DOI: 10.1177/0271678x18808174] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ischemic injury can be alleviated by the judicious use of hypothermia. However, the optimal regimens and the temporal kinetics of post-stroke neurovascular responses to hypothermic intervention have not been systematically studied. These gaps slow the clinical translation of hypothermia as an anti-stroke therapy. Here, we characterized the effects of transient selective brain hypothermia (TSBH) from the hyperacute to chronic stages of focal ischemia/reperfusion brain injury induced by transient middle cerebral artery occlusion in mice. A simple cooling device was used to induce TSBH during cerebral ischemia. This treatment reduced mortality from 31.8% to 0% and improved neurological outcomes for at least 35 days post-injury. TSBH mitigated blood-brain barrier leakage during the hyperacute and acute injury stages (1-23 h post-reperfusion). This early protection of the blood-brain barrier was associated with anti-inflammatory phenotypic polarization of microglia/macrophages, reduced production of pro-inflammatory cytokines, and less brain infiltration of neutrophils and macrophages during the subacute injury stage (three days post-reperfusion). TSBH elicited enduring protective effects on both grey and white matter for at least 35 days post-injury and preserved the long-term electrophysiological function of fiber tracts. In conclusion, TSBH ameliorates ischemia/reperfusion injury in the neurovascular unit from hyperacute to chronic injury stages after experimental stroke.
Collapse
Affiliation(s)
- Jingyan Zhao
- 1 Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,2 Stroke Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,3 Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hongfeng Mu
- 3 Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Liqiang Liu
- 2 Stroke Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,3 Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoyan Jiang
- 3 Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Di Wu
- 1 Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yejie Shi
- 3 Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rehana K Leak
- 4 Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Xunming Ji
- 1 Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,2 Stroke Center, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.,5 Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
61
|
Stoll G, Nieswandt B. Thrombo-inflammation in acute ischaemic stroke — implications for treatment. Nat Rev Neurol 2019; 15:473-481. [DOI: 10.1038/s41582-019-0221-1] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2019] [Indexed: 01/17/2023]
|
62
|
Malone K, Amu S, Moore AC, Waeber C. Immunomodulatory Therapeutic Strategies in Stroke. Front Pharmacol 2019; 10:630. [PMID: 31281252 PMCID: PMC6595144 DOI: 10.3389/fphar.2019.00630] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/16/2019] [Indexed: 12/14/2022] Open
Abstract
The role of immunity in all stages of stroke is increasingly being recognized, from the pathogenesis of risk factors to tissue repair, leading to the investigation of a range of immunomodulatory therapies. In the acute phase of stroke, proposed therapies include drugs targeting pro-inflammatory cytokines, matrix metalloproteinases, and leukocyte infiltration, with a key objective to reduce initial brain cell toxicity. Systemically, the early stages of stroke are also characterized by stroke-induced immunosuppression, where downregulation of host defences predisposes patients to infection. Therefore, strategies to modulate innate immunity post-stroke have garnered greater attention. A complementary objective is to reduce longer-term sequelae by focusing on adaptive immunity. Following stroke onset, the integrity of the blood–brain barrier is compromised, exposing central nervous system (CNS) antigens to systemic adaptive immune recognition, potentially inducing autoimmunity. Some pre-clinical efforts have been made to tolerize the immune system to CNS antigens pre-stroke. Separately, immune cell populations that exhibit a regulatory phenotype (T- and B- regulatory cells) have been shown to ameliorate post-stroke inflammation and contribute to tissue repair. Cell-based therapies, established in oncology and transplantation, could become a strategy to treat the acute and chronic stages of stroke. Furthermore, a role for the gut microbiota in ischaemic injury has received attention. Finally, the immune system may play a role in remote ischaemic preconditioning-mediated neuroprotection against stroke. The development of stroke therapies involving organs distant to the infarct site, therefore, should not be overlooked. This review will discuss the immune mechanisms of various therapeutic strategies, surveying published data and discussing more theoretical mechanisms of action that have yet to be exploited.
Collapse
Affiliation(s)
- Kyle Malone
- Department of Pharmacology and Therapeutics, School of Pharmacy, University College Cork, Cork, Ireland
| | - Sylvie Amu
- Cancer Research @UCC, University College Cork, Cork, Ireland
| | - Anne C Moore
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Christian Waeber
- Department of Pharmacology and Therapeutics, School of Pharmacy, University College Cork, Cork, Ireland
| |
Collapse
|
63
|
Sîrbulescu RF, Chung JY, Edmiston WJ, Poznansky SA, Poznansky MC, Whalen MJ. Intraparenchymal Application of Mature B Lymphocytes Improves Structural and Functional Outcome after Contusion Traumatic Brain Injury. J Neurotrauma 2019; 36:2579-2589. [PMID: 30997843 DOI: 10.1089/neu.2018.6368] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cerebral contusion causes neurological dysfunction mediated in part by inflammatory responses to injury. B lymphocytes are dynamic regulators of the immune system that have not been systematically studied in traumatic brain injury (TBI). We showed previously that topically applied mature B cells have immunomodulatory properties and strongly promote tissue regeneration, including cutaneous nerve growth, in acute and chronic skin wounds. Using a mouse controlled cortical impact (CCI) model, we assessed a possible beneficial role of exogenously applied B cells on histopathological and functional outcome after TBI. Mice were injected intraparenchymally at the lesion site with 2 × 106 mature naïve syngeneic splenic B cells, then subjected to CCI. Control CCI mice received equal numbers of T cells or saline, and sham-injured mice (craniotomy only) were given B cells or saline. Sham-injured groups performed similarly in motor and learning tests. Injured mice administered B cells showed significantly improved post-injury rotarod, Y maze, and Morris water maze (MWM) performance compared with saline- or T-cell-treated CCI groups. Moreover, lesion volume in mice treated with B cells was significantly reduced by 40% at 35 days post-TBI compared with saline and T cell controls, and astrogliosis and microglial activation were decreased. In vivo tracking of exogenous B cells showed that they have a limited life span of approximately 14 days in situ and do not appear to proliferate. The data suggest proof of principle that local administration of B lymphocytes may represent a therapeutic option for treatment of cerebral contusion, especially when clinical management involves procedures that allow access to the injury site.
Collapse
Affiliation(s)
- Ruxandra F Sîrbulescu
- Vaccine and Immunotherapy Center, Department of Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Joon Yong Chung
- Neuroscience Center, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - William J Edmiston
- Neuroscience Center, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sonya A Poznansky
- Neuroscience Center, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Department of Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael J Whalen
- Neuroscience Center, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
64
|
Javidi E, Magnus T. Autoimmunity After Ischemic Stroke and Brain Injury. Front Immunol 2019; 10:686. [PMID: 31001280 PMCID: PMC6454865 DOI: 10.3389/fimmu.2019.00686] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/13/2019] [Indexed: 12/20/2022] Open
Abstract
Ischemic Stroke is a major cause of morbidity and mortality worldwide. Sterile inflammation occurs after both stroke subtypes and contributes to neuronal injury and damage to the blood-brain barrier with release of brain antigens and a potential induction of autoimmune responses that escape central and peripheral tolerance mechanisms. In stroke patients, the detection of T cells and antibodies specific to neuronal antigens suggests a role of humoral adaptive immunity. In experimental models stroke leads to a significant increase of autoreactive T and B cells to CNS antigens. Lesion volume and functional outcome in stroke patients and murine stroke models are connected to antigen-specific responses to brain proteins. In patients with traumatic brain injury (TBI) a range of antibodies against brain proteins can be detected in serum samples. In this review, we will summarize the role of autoimmunity in post-lesional conditions and discuss the role of B and T cells and their potential neuroprotective or detrimental effects.
Collapse
Affiliation(s)
- Ehsan Javidi
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
65
|
Wang Z, He D, Zeng YY, Zhu L, Yang C, Lu YJ, Huang JQ, Cheng XY, Huang XH, Tan XJ. The spleen may be an important target of stem cell therapy for stroke. J Neuroinflammation 2019; 16:20. [PMID: 30700305 PMCID: PMC6352449 DOI: 10.1186/s12974-019-1400-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 01/07/2019] [Indexed: 12/21/2022] Open
Abstract
Stroke is the most common cerebrovascular disease, the second leading cause of death behind heart disease and is a major cause of long-term disability worldwide. Currently, systemic immunomodulatory therapy based on intravenous cells is attracting attention. The immune response to acute stroke is a major factor in cerebral ischaemia (CI) pathobiology and outcomes. Over the past decade, the significant contribution of the spleen to ischaemic stroke has gained considerable attention in stroke research. The changes in the spleen after stroke are mainly reflected in morphology, immune cells and cytokines, and these changes are closely related to the stroke outcomes. Autonomic nervous system (ANS) activation, release of central nervous system (CNS) antigens and chemokine/chemokine receptor interactions have been documented to be essential for efficient brain-spleen cross-talk after stroke. In various experimental models, human umbilical cord blood cells (hUCBs), haematopoietic stem cells (HSCs), bone marrow stem cells (BMSCs), human amnion epithelial cells (hAECs), neural stem cells (NSCs) and multipotent adult progenitor cells (MAPCs) have been shown to reduce the neurological damage caused by stroke. The different effects of these cell types on the interleukin (IL)-10, interferon (IFN), and cholinergic anti-inflammatory pathways in the spleen after stroke may promote the development of new cell therapy targets and strategies. The spleen will become a potential target of various stem cell therapies for stroke represented by MAPC treatment.
Collapse
Affiliation(s)
- Zhe Wang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China.,Institute of Reproductive and Stem Cell Research, School of Basic Medical Science, Central South University, Changsha, 410000, China
| | - Da He
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Ya-Yue Zeng
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Li Zhu
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Chao Yang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Yong-Juan Lu
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Jie-Qiong Huang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiao-Yan Cheng
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiang-Hong Huang
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China
| | - Xiao-Jun Tan
- Xiangtan Central Hospital, Clinical Practice Base of Central South University, Xiangtan, 411100, China.
| |
Collapse
|
66
|
Co-signaling Molecules in Neurological Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:233-265. [DOI: 10.1007/978-981-32-9717-3_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
67
|
Yang C, Hawkins KE, Doré S, Candelario-Jalil E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol 2018; 316:C135-C153. [PMID: 30379577 DOI: 10.1152/ajpcell.00136.2018] [Citation(s) in RCA: 481] [Impact Index Per Article: 80.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
As part of the neurovascular unit, the blood-brain barrier (BBB) is a unique, dynamic regulatory boundary that limits and regulates the exchange of molecules, ions, and cells between the blood and the central nervous system. Disruption of the BBB plays an important role in the development of neurological dysfunction in ischemic stroke. Blood-borne substances and cells have restricted access to the brain due to the presence of tight junctions between the endothelial cells of the BBB. Following stroke, there is loss of BBB tight junction integrity, leading to increased paracellular permeability, which results in vasogenic edema, hemorrhagic transformation, and increased mortality. Thus, understanding principal mediators and molecular mechanisms involved in BBB disruption is critical for the development of novel therapeutics to treat ischemic stroke. This review discusses the current knowledge of how neuroinflammation contributes to BBB damage in ischemic stroke. Specifically, we provide an updated overview of the role of cytokines, chemokines, oxidative and nitrosative stress, adhesion molecules, matrix metalloproteinases, and vascular endothelial growth factor as well as the role of different cell types in the regulation of BBB permeability in ischemic stroke.
Collapse
Affiliation(s)
- Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Kimberly E Hawkins
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Sylvain Doré
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida.,Departments of Anesthesiology, Neurology, Psychiatry, Psychology, and Pharmaceutics, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| |
Collapse
|
68
|
Wimmer I, Zrzavy T, Lassmann H. Neuroinflammatory responses in experimental and human stroke lesions. J Neuroimmunol 2018; 323:10-18. [DOI: 10.1016/j.jneuroim.2018.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 02/07/2023]
|
69
|
Hu X, Leak RK, Thomson AW, Yu F, Xia Y, Wechsler LR, Chen J. Promises and limitations of immune cell-based therapies in neurological disorders. Nat Rev Neurol 2018; 14:559-568. [PMID: 29925925 PMCID: PMC6237550 DOI: 10.1038/s41582-018-0028-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The healthy immune system has natural checkpoints that temper pernicious inflammation. Cells mediating these checkpoints include regulatory T cells, regulatory B cells, regulatory dendritic cells, microglia, macrophages and monocytes. Here, we highlight discoveries on the beneficial functions of regulatory immune cells and their mechanisms of action and evaluate their potential use as novel cell-based therapies for brain disorders. Regulatory immune cell therapies have the potential not only to mitigate the exacerbation of brain injury by inflammation but also to promote an active post-injury brain repair programme. By harnessing the reparative properties of these cells, we can reduce over-reliance on medications that mask clinical symptoms but fail to impede or reverse the progression of brain disorders. Although these discoveries encourage further testing and genetic engineering of regulatory immune cells for the clinical management of neurological disorders, a number of challenges must be surmounted to improve their safety and efficacy in humans.
Collapse
Affiliation(s)
- Xiaoming Hu
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Angus W Thomson
- Starzl Transplantation Institute, Department of Surgery and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Fang Yu
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yuguo Xia
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lawrence R Wechsler
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders and Recovery and Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
70
|
An S, Wang X, Ruck MA, Rodriguez HJ, Kostyushev DS, Varga M, Luu E, Derakhshandeh R, Suchkov SV, Kogan SC, Hermiston ML, Springer ML. Age-Related Impaired Efficacy of Bone Marrow Cell Therapy for Myocardial Infarction Reflects a Decrease in B Lymphocytes. Mol Ther 2018; 26:1685-1693. [PMID: 29914756 DOI: 10.1016/j.ymthe.2018.05.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 12/19/2022] Open
Abstract
Treatment of myocardial infarction (MI) with bone marrow cells (BMCs) improves post-MI cardiac function in rodents. However, clinical trials of BMC therapy have been less effective. While most rodent experiments use young healthy donors, patients undergoing autologous cell therapy are older and post-MI. We previously demonstrated that BMCs from aged and post-MI donor mice are therapeutically impaired, and that donor MI induces inflammatory changes in BMC composition including reduced levels of B lymphocytes. Here, we hypothesized that B cell alterations in bone marrow account for the reduced therapeutic potential of post-MI and aged donor BMCs. Injection of BMCs from increasingly aged donor mice resulted in progressively poorer cardiac function and larger infarct size. Flow cytometry revealed fewer B cells in aged donor bone marrow. Therapeutic efficacy of young healthy donor BMCs was reduced by depletion of B cells. Implantation of intact or lysed B cells improved cardiac function, whereas intact or lysed T cells provided only minor benefit. We conclude that B cells play an important paracrine role in effective BMC therapy for MI. Reduction of bone marrow B cells because of age or MI may partially explain why clinical autologous cell therapy has not matched the success of rodent experiments.
Collapse
Affiliation(s)
- Songtao An
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Division of Cardiology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Xiaoyin Wang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Melissa A Ruck
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hilda J Rodriguez
- Division of Cardiology, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dmitry S Kostyushev
- Division of Cardiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Monika Varga
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Emmy Luu
- Division of Cardiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ronak Derakhshandeh
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sergey V Suchkov
- Center for Personalized Medicine, Sechenov University, Moscow, Russia; Department for Translational Medicine, Moscow Engineering Physical Institute, Moscow, Russia
| | - Scott C Kogan
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michelle L Hermiston
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew L Springer
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA; Division of Cardiology, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
71
|
Chen C, Jiang W, Liu Z, Li F, Yang J, Zhao Y, Ran Y, Meng Y, Ji X, Geng X, Du H, Hu X. Splenic responses play an important role in remote ischemic preconditioning-mediated neuroprotection against stroke. J Neuroinflammation 2018; 15:167. [PMID: 29807548 PMCID: PMC5972448 DOI: 10.1186/s12974-018-1190-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 05/06/2018] [Indexed: 12/24/2022] Open
Abstract
Background Remote ischemic preconditioning (RIPC) of a limb has been reported to protect against ischemic stroke. Our previous results demonstrated that the RIPC-mediated neuroprotection is associated with alterations in circulating immune cell populations. Here, we evaluated the effect of the spleen, the largest reservoir of immune cells, on RIPC-mediated neuroprotection against stroke. Methods Noninvasive RIPC was achieved by four repeated cycles of 5-min blood flow constriction in the hindlimbs using a tourniquet. The blood and spleens were collected before and 1 h and 3 days after preconditioning to analyze the effect of RIPC on the spleen and the correlation between splenic and peripheral lymphocytes. Moreover, spleen weight and splenic lymphocytes were compared in stroke rats with or without RIPC. Finally, splenectomy was made 1 day or 2 weeks before RIPC and 90-min middle cerebral artery occlusion (MCAO). The infarct areas and deficits were assessed. Blood was collected 1 h after RIPC and 3 days after MCAO to explore the impact of splenectomy on RIPC-induced neuroprotection and immune changes. The contralateral and ipsilateral hemispheres were collected 3 days after MCAO to detect the infiltration of immune cells after RIPC and splenectomy. Results Flow cytometry analysis demonstrated that the RIPC promptly increased the percentages of CD3+CD8+ cytotoxic T (Tc) cells in the spleen with a relatively delayed elevation in CD3+CD161+ natural killer T (NKT) and CD3−CD45RA+ B lymphocytes. The percentages of circulating lymphocytes are positively correlated with the percentages of splenic lymphocytes in normal rats. Interestingly, RIPC resulted in negative correlations between the percentages of splenic and circulating T lymphocytes, while the correlation between splenic and circulating B lymphocytes remained positive. For animals subjected to RIPC followed by MCAO, RIPC increased splenic volume with an expansion of splenic lymphocytes 3 days after MCAO. Furthermore, the removal of the spleen 1 day or 2 weeks before RIPC and MCAO reduced the protective effect of RIPC on ischemic brain injury and reversed the effects of RIPC on circulating immune cell composition. RIPC significantly reduced brain infiltration of Tc and NKT cells. Prior splenectomy showed no effect on immune cell infiltration after RIPC and stroke. Conclusion These results reveal an immunomodulatory effect of the spleen, effecting mainly the spleen-derived lymphocytes, during RIPC-afforded neuroprotection against cerebral ischemia. Electronic supplementary material The online version of this article (10.1186/s12974-018-1190-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chen Chen
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China
| | - Wei Jiang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China
| | - Zongjian Liu
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China
| | - Jian Yang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China
| | - Yanlong Zhao
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China
| | - Yuanyuan Ran
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China
| | - Yan Meng
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xunming Ji
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China.,Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China
| | - Huishan Du
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China.
| | - Xiaoming Hu
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, 101100, China.
| |
Collapse
|
72
|
Talhada D, Rabenstein M, Ruscher K. The role of dopaminergic immune cell signalling in poststroke inflammation. Ther Adv Neurol Disord 2018; 11:1756286418774225. [PMID: 29774058 PMCID: PMC5952273 DOI: 10.1177/1756286418774225] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/06/2018] [Indexed: 01/08/2023] Open
Abstract
Upon ischaemic stroke, brain-resident and peripheral immune cells accumulate in the central nervous system (CNS). Interestingly, these cells express pattern specific to neurotransmitter receptors and, therefore, seem to be susceptible to neurotransmitter stimulation, potentially modulating their properties and functions. One of the principal neurotransmitters in the CNS, dopamine, is involved in the regulation of processes of brain development, motor control and higher brain functions. It is constantly released in the brain and there is experimental and clinical evidence that dopaminergic signalling is involved in recovery of lost neurological function after stroke. Independent studies have revealed specific but different patterns of dopamine receptor subtypes on different populations of immune cells. Those patterns are dependent on the activation status of cells. Generally, exposure to dopamine or dopamine receptor agonists decreases detrimental actions of immune cells. In contrast, a reduction of dopaminergic inputs perpetuates a pro-inflammatory state associated with increased release of pro-inflammatory molecules. In addition, subsets of immune cells have been identified to synthesize and release dopamine, suggesting autoregulatory mechanisms. Evidence supports that inflammatory processes activated following ischaemic stroke are modulated by dopaminergic signalling.
Collapse
Affiliation(s)
- Daniela Talhada
- LUBIN Lab – Lund Brain Injury Laboratory for Neurosurgical Research, Department of Clinical Sciences, Lund University, Lund, Sweden CICS-UBI-Health Sciences Research Centre, Faculdade de Ciências da Saúde, Av. Infante D. Henrique, Universidade da Beira Interior, Portugal
| | - Monika Rabenstein
- Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Karsten Ruscher
- Lund Brain Injury Laboratory for Neurosurgical Research, Wallenberg Neuroscience Center, Lund University, BMC A13, S-22184 Lund, Sweden
| |
Collapse
|
73
|
Coder B, Wang W, Wang L, Wu Z, Zhuge Q, Su DM. Friend or foe: the dichotomous impact of T cells on neuro-de/re-generation during aging. Oncotarget 2018; 8:7116-7137. [PMID: 27738345 PMCID: PMC5351694 DOI: 10.18632/oncotarget.12572] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/05/2016] [Indexed: 12/15/2022] Open
Abstract
The interaction between T cells and the central nervous system (CNS) in homeostasis and injury has been recognized being both pathogenic (CD4+ T-helper 1 - Th1, Th17 and γδT) and ameliorative (Th2 and regulatory T cells - Tregs). However, in-depth studies aimed to elucidate the precise in the aged microenvironment and the dichotomous role of Tregs have just begun and many aspects remain unclear. This is due, not only to a mutual dependency and reciprocal causation of alterations and diseases between the nervous and T cell immune systems, but also to an inconsistent aging of the two systems, which dynamically changes with CNS injury/recovery and/or aging process. Cellular immune system aging, particularly immunosenescence and T cell aging initiated by thymic involution - sources of chronic inflammation in the elderly (termed inflammaging), potentially induces an acceleration of brain aging and memory loss. In turn, aging of the brain via neuro-endocrine-immune network drives total body systemic aging, including that of the immune system. Therefore, immunotherapeutics including vaccination and “protective autoimmunity” provide promising means to rejuvenate neuro-inflammatory disorders and repair CNS acute injury and chronic neuro-degeneration. We review the current understanding and recent discoveries linking the aging immune system with CNS injury and neuro-degeneration. Additionally, we discuss potential recovery and rejuvenation strategies, focusing on targeting the aging T cell immune system in an effort to alleviate acute brain injury and chronic neuro-degeneration during aging, via the “thymus-inflammaging-neurodegeneration axis”.
Collapse
Affiliation(s)
- Brandon Coder
- Institute of Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Weikan Wang
- Institute of Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang, P. R. China
| | - Liefeng Wang
- Institute of Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA.,Department of Biotechnology, Gannan Medical University, Ganzhou, P. R. China
| | - Zhongdao Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, P. R. China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang, P. R. China
| | - Dong-Ming Su
- Institute of Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
74
|
Seifert HA, Vandenbark AA, Offner H. Regulatory B cells in experimental stroke. Immunology 2018; 154:169-177. [PMID: 29313944 DOI: 10.1111/imm.12887] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/21/2017] [Accepted: 12/28/2017] [Indexed: 12/30/2022] Open
Abstract
Current treatment options for human stroke are limited mainly to the modestly effective infusion of tissue plasminogen activator (tPA), with additional improvement of functional independence and higher rates of angiographic revascularization observed after mechanical thrombectomy. However, new therapeutic strategies that address post-stroke immune-mediated inflammatory responses are urgently needed. Recent studies in experimental stroke have firmly implicated immune mechanisms in the propagation and partial resolution of central nervous system damage after the ischaemic event. A new-found anti-inflammatory role for regulatory B (Breg) cells in autoimmune diseases sparked interest in these cells as potential immunomodulators in stroke. Subsequent studies identified interleukin-10 as a common regulatory cytokine among all five of the currently recognized Breg cell subsets, several of which can be found in the affected brain hemisphere after induction of experimental stroke in mice. Transfer of enriched Breg cell subpopulations into both B-cell-depleted and wild-type mice confirmed their potent immunosuppressive activities in vivo, including recruitment and potentiation of regulatory T cells. Moreover, Breg cell therapy strongly reduced stroke volumes and treatment outcomes in ischaemic mice even when administered 24 hr after induction of experimental stroke, a treatment window far exceeding that of tPA. These striking results suggest that transfer of enriched Breg cell populations could have therapeutic value in human stroke, although considerable clinical challenges remain.
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
| | - 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
| | - 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
|
75
|
Pivotal neuroinflammatory and therapeutic role of high mobility group box 1 in ischemic stroke. Biosci Rep 2017; 37:BSR20171104. [PMID: 29054968 PMCID: PMC5715129 DOI: 10.1042/bsr20171104] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/29/2017] [Accepted: 10/18/2017] [Indexed: 12/27/2022] Open
Abstract
Stroke is a major cause of mortality and disability worldwide. Stroke is a frequent and severe neurovascular disorder. The main cause of stroke is atherosclerosis, and the most common risk factor for atherosclerosis is hypertension. Therefore, prevention and treatment of stroke are crucial issues in humans. High mobility group box 1 (HMGB1) is non-histone nuclear protein that is currently one of the crucial proinflammatory alarmins in ischemic stroke (IS). It is instantly released from necrotic cells in the ischemic core and activates an early inflammatory response. HMGB1 may signal via its putative receptors, such as receptor for advanced glycation end products (RAGE), toll-like receptors (TLRs) as well as matrix metalloproteinase (MMP) enzymes during IS. These receptors are expressed in brain cells. Additionally, brain-released HMGB1 can be redox modified in the circulation and activate peripheral immune cells. The role of HMGB1 may be more complex. HMGB1 possesses beneficial actions, such as endothelial activation, enhancement of neurite outgrowth, and neuronal survival. HMGB1 may also provide a novel link for brain-immune communication leading to post-stroke immunomodulation. Therefore, HMGB1 is new promising therapeutic intervention aimed at promoting neurovascular repair and remodeling after stroke. In this review, we look at the mechanisms of secretion of HMGB1, the role of receptors, MMP enzymes, hypoglycemia, atherosclerosis, edema, angiogenesis as well as neuroimmunological reactions and post-ischemic brain recovery in IS. We also outline therapeutic roles of HMGB1 in IS.
Collapse
|
76
|
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: 144] [Impact Index Per Article: 20.6] [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
|
77
|
Impact of aging immune system on neurodegeneration and potential immunotherapies. Prog Neurobiol 2017; 157:2-28. [PMID: 28782588 DOI: 10.1016/j.pneurobio.2017.07.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 07/25/2017] [Accepted: 07/28/2017] [Indexed: 12/19/2022]
Abstract
The interaction between the nervous and immune systems during aging is an area of avid interest, but many aspects remain unclear. This is due, not only to the complexity of the aging process, but also to a mutual dependency and reciprocal causation of alterations and diseases between both the nervous and immune systems. Aging of the brain drives whole body systemic aging, including aging-related changes of the immune system. In turn, the immune system aging, particularly immunosenescence and T cell aging initiated by thymic involution that are sources of chronic inflammation in the elderly (termed inflammaging), potentially induces brain aging and memory loss in a reciprocal manner. Therefore, immunotherapeutics including modulation of inflammation, vaccination, cellular immune therapies and "protective autoimmunity" provide promising approaches to rejuvenate neuroinflammatory disorders and repair brain injury. In this review, we summarize recent discoveries linking the aging immune system with the development of neurodegeneration. Additionally, we discuss potential rejuvenation strategies, focusing aimed at targeting the aging immune system in an effort to prevent acute brain injury and chronic neurodegeneration during aging.
Collapse
|
78
|
Nakajima M, Nito C, Sowa K, Suda S, Nishiyama Y, Nakamura-Takahashi A, Nitahara-Kasahara Y, Imagawa K, Hirato T, Ueda M, Kimura K, Okada T. Mesenchymal Stem Cells Overexpressing Interleukin-10 Promote Neuroprotection in Experimental Acute Ischemic Stroke. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 6:102-111. [PMID: 28725658 PMCID: PMC5502709 DOI: 10.1016/j.omtm.2017.06.005] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/21/2017] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-10 is a contributing factor to neuroprotection of mesenchymal stem cell (MSC) transplantation after ischemic stroke. Our aim was to increase therapeutic effects by combining MSCs and ex vivo IL-10 gene transfer with an adeno-associated virus (AAV) vector using a rat transient middle cerebral artery occlusion (MCAO) model. Sprague-Dawley rats underwent 90 min MCAO followed by intravenous administration of MSCs alone or IL-10 gene-transferred MSCs (MSC/IL-10) at 0 or 3 hr after ischemia reperfusion. Infarct lesions, neurological deficits, and immunological analyses were performed within 7 days after MCAO. 0-hr transplantation of MSCs alone and MSC/IL-10 significantly reduced infarct volumes and improved motor function. Conversely, 3-hr transplantation of MSC/IL-10, but not MSCs alone, significantly reduced infarct volumes (p < 0.01) and improved motor function (p < 0.01) compared with vehicle groups at 72 hr and 7 days after MCAO. Immunological analysis showed that MSC/IL-10 transplantation significantly inhibits microglial activation and pro-inflammatory cytokine expression compared with MSCs alone. Moreover, overexpressing IL-10 suppressed neuronal degeneration and improved survival of engrafted MSCs in the ischemic hemisphere. These results suggest that overexpressing IL-10 enhances the neuroprotective effects of MSC transplantation by anti-inflammatory modulation and thereby supports neuronal survival during the acute ischemic phase.
Collapse
Affiliation(s)
- Masataka Nakajima
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Chikako Nito
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Kota Sowa
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Satoshi Suda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Yasuhiro Nishiyama
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Aki Nakamura-Takahashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Pharmacology, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Yuko Nitahara-Kasahara
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Cell and Gene Therapy, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | | | - Tohru Hirato
- JCR Pharmaceuticals Co., Ltd., Hyogo, 659-0021, Japan
| | - Masayuki Ueda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Kazumi Kimura
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan.,Department of Cell and Gene Therapy, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8603, Japan
| |
Collapse
|
79
|
Garcia JM, Stillings SA, Leclerc JL, Phillips H, Edwards NJ, Robicsek SA, Hoh BL, Blackburn S, Doré S. Role of Interleukin-10 in Acute Brain Injuries. Front Neurol 2017; 8:244. [PMID: 28659854 PMCID: PMC5466968 DOI: 10.3389/fneur.2017.00244] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/17/2017] [Indexed: 12/23/2022] Open
Abstract
Interleukin-10 (IL-10) is an important anti-inflammatory cytokine expressed in response to brain injury, where it facilitates the resolution of inflammatory cascades, which if prolonged causes secondary brain damage. Here, we comprehensively review the current knowledge regarding the role of IL-10 in modulating outcomes following acute brain injury, including traumatic brain injury (TBI) and the various stroke subtypes. The vascular endothelium is closely tied to the pathophysiology of these neurological disorders and research has demonstrated clear vascular endothelial protective properties for IL-10. In vitro and in vivo models of ischemic stroke have convincingly directly and indirectly shown IL-10-mediated neuroprotection; although clinically, the role of IL-10 in predicting risk and outcomes is less clear. Comparatively, conclusive studies investigating the contribution of IL-10 in subarachnoid hemorrhage are lacking. Weak indirect evidence supporting the protective role of IL-10 in preclinical models of intracerebral hemorrhage exists; however, in the limited number of clinical studies, higher IL-10 levels seen post-ictus have been associated with worse outcomes. Similarly, preclinical TBI models have suggested a neuroprotective role for IL-10; although, controversy exists among the several clinical studies. In summary, while IL-10 is consistently elevated following acute brain injury, the effect of IL-10 appears to be pathology dependent, and preclinical and clinical studies often paradoxically yield opposite results. The pronounced and potent effects of IL-10 in the resolution of inflammation and inconsistency in the literature regarding the contribution of IL-10 in the setting of acute brain injury warrant further rigorously controlled and targeted investigation.
Collapse
Affiliation(s)
- Joshua M Garcia
- College of Medicine, University of Florida, Gainesville, FL, United States
| | | | - Jenna L Leclerc
- Department of Anesthesiology, College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States.,Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Harrison Phillips
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States
| | - Nancy J Edwards
- Department of Neurology, University of California, San Francisco, CA, United States.,Department of Neurosurgery, University of California, San Francisco, CA, United States
| | - Steven A Robicsek
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States.,Department of Neurosurgery, University of Florida, Gainesville, FL, United States.,Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Brian L Hoh
- Department of Neurosurgery, University of Florida, Gainesville, FL, United States
| | - Spiros Blackburn
- Department of Neurosurgery, University of Texas, Houston, TX, United States
| | - Sylvain Doré
- Department of Anesthesiology, College of Medicine, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States.,Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, United States.,Department of Neurology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Department of Psychology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Department of Psychiatry, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Department of Pharmaceutics, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
80
|
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: 22] [Impact Index Per Article: 3.1] [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
|
81
|
Selvaraj UM, Ortega SB, Hu R, Gilchrist R, Kong X, Partin A, Plautz EJ, Klein RS, Gidday JM, Stowe AM. Preconditioning-induced CXCL12 upregulation minimizes leukocyte infiltration after stroke in ischemia-tolerant mice. J Cereb Blood Flow Metab 2017; 37:801-813. [PMID: 27006446 PMCID: PMC5363460 DOI: 10.1177/0271678x16639327] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Repetitive hypoxic preconditioning creates long-lasting, endogenous protection in a mouse model of stroke, characterized by reductions in leukocyte-endothelial adherence, inflammation, and infarct volumes. The constitutively expressed chemokine CXCL12 can be upregulated by hypoxia and limits leukocyte entry into brain parenchyma during central nervous system inflammatory autoimmune disease. We therefore hypothesized that the sustained tolerance to stroke induced by repetitive hypoxic preconditioning is mediated, in part, by long-term CXCL12 upregulation at the blood-brain barrier (BBB). In male Swiss Webster mice, repetitive hypoxic preconditioning elevated cortical CXCL12 protein levels, and the number of cortical CXCL12+ microvessels, for at least two weeks after the last hypoxic exposure. Repetitive hypoxic preconditioning-treated mice maintained more CXCL12-positive vessels than untreated controls following transient focal stroke, despite cortical decreases in CXCL12 mRNA and protein. Continuous administration of the CXCL12 receptor (CXCR4) antagonist AMD3100 for two weeks following repetitive hypoxic preconditioning countered the increase in CXCL12-positive microvessels, both prior to and following stroke. AMD3100 blocked the protective post-stroke reductions in leukocyte diapedesis, including macrophages and NK cells, and blocked the protective effect of repetitive hypoxic preconditioning on lesion volume, but had no effect on blood-brain barrier dysfunction. These data suggest that CXCL12 upregulation prior to stroke onset, and its actions following stroke, contribute to the endogenous, anti-inflammatory phenotype induced by repetitive hypoxic preconditioning.
Collapse
Affiliation(s)
- Uma Maheswari Selvaraj
- 1 Department of Neurology & Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sterling B Ortega
- 1 Department of Neurology & Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ruilong Hu
- 2 Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert Gilchrist
- 2 Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiangmei Kong
- 1 Department of Neurology & Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Alexander Partin
- 1 Department of Neurology & Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Erik J Plautz
- 1 Department of Neurology & Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Robyn S Klein
- 4 Department of Medicine, Washington University, St Louis, MO, USA
| | - Jeffrey M Gidday
- 2 Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA.,3 Department of Ophthalmology, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Ann M Stowe
- 1 Department of Neurology & Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
82
|
Role of interleukin-10 in the neuroprotective effect of the Angiotensin Type 2 Receptor agonist, compound 21, after ischemia/reperfusion injury. Eur J Pharmacol 2017; 799:128-134. [PMID: 28192099 DOI: 10.1016/j.ejphar.2017.02.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 01/04/2023]
Abstract
INTRODUCTION We and others have shown that the angiotensin type 2 (AT2) receptor agonist, compound 21 (C21), provides neuroprotection and enhances recovery in rodent stroke models yet the mechanism involved is not known. Moreover, C21 treatment is associated with an anti-inflammatory response. Here we tested the hypothesis that C21 mediates neuroprotection by upregulating the neuroprotective and anti-inflammatory cytokine, interleukin (IL)-10. METHODS Wistar rats were subjected to 3h-middle cerebral artery suture occlusion and treated at reperfusion with C21 (0.03mg/kg)±IL-10 neutralizing antibody (0.1mg/kg) both given i.p. Infarct size, behavioral outcomes, and molecular analysis were performed at 24h post-injury. Primary rat neurons were used to test the direct neuroprotective effect of C21 in vitro. RESULTS C21 treatment reduced infarct size, improved functional outcome and decreased the pro-inflammatory cytokine, tumor necrosis factor alpha (TNF-α) in the ischemic hemisphere compared to saline. Anti-IL-10 co-treatment blocked the C21-induced reduction in infarct size and inflammation, and the improvement in behavioral outcome. In vitro, C21 treatment increased neuron survival and reduced cell apoptosis after oxygen glucose deprivation (OGD) and OGD/reoxygenation. These effects were mediated through AT2R stimulation. CONCLUSION C21 provides direct neuroprotection as well as indirect protection through IL-10.
Collapse
|
83
|
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: 40] [Impact Index Per Article: 5.0] [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
|
84
|
Sun N, Keep RF, Hua Y, Xi G. Critical Role of the Sphingolipid Pathway in Stroke: a Review of Current Utility and Potential Therapeutic Targets. Transl Stroke Res 2016; 7:420-38. [PMID: 27339463 DOI: 10.1007/s12975-016-0477-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/12/2016] [Accepted: 06/15/2016] [Indexed: 12/16/2022]
Abstract
Sphingolipids are a series of cell membrane-derived lipids which act as signaling molecules and play a critical role in cell death and survival, proliferation, recognition, and migration. Sphingosine-1-phosphate acts as a key signaling molecule and regulates lymphocyte trafficking, glial cell activation, vasoconstriction, endothelial barrier function, and neuronal death pathways which plays a critical role in numerous neurological conditions. Stroke is a second leading cause of death all over the world and effective therapies are still in great demand, including ischemic stroke and hemorrhagic stroke as well as poststroke repair. Significantly, sphingolipid activities change after stroke and correlate with stroke outcome, which has promoted efforts to testify whether the sphingolipid pathway could be a novel therapeutic target in stroke. The sphingolipid metabolic pathway, the connection between the pathway and stroke, as well as therapeutic interventions to manipulate the pathway to reduce stroke-induced brain injury are discussed in this review.
Collapse
Affiliation(s)
- Na Sun
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
85
|
Ji M, Wang Y, Wang L, Chen L, Li J. Protective effect of Xuebijing injection against acute lung injury induced by left ventricular ischemia/reperfusion in rabbits. Exp Ther Med 2016; 12:51-58. [PMID: 27347017 PMCID: PMC4906817 DOI: 10.3892/etm.2016.3275] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/19/2016] [Indexed: 12/11/2022] Open
Abstract
Xuebijing (XBJ) is a Chinese herbal preparation. Previous studies have demonstrated that XBJ injection is able to inhibit the uncontrolled release of endogenous inflammatory mediators, attenuate inflammation, and alleviate organ damage. However, there are no relevant reports on the protective effect of XBJ against left ventricular ischemia/reperfusion (I/R)-induced acute lung injury (ALI). Therefore, the aim of the present study was to evaluate the protective effect of XBJ on ALI induced by left ventricular I/R, and provide evidence for the clinical application of XBJ. In the present study, 120 healthy rabbits of mixed gender were randomly assigned to a normal control group, ischemia group, I/R group (I/RG) and XBJ-injection treatment group (TG). In addition, each group was further divided into three subgroups (n=10/subgroup), namely, 30 min pre-ischemia, 30 min post-ischemia and 30 min post-reperfusion subgroups. Blood samples (5 ml) were collected from the jugularis externa and carotis communis of the rabbits at the three time points, and a blood gas analyzer was used to measure the arterial partial pressure of oxygen (PaO2) and carbon dioxide (PaCO2). Following sacrifice, the lungs of the rabbits were removed and a bronchoalveolar lavage (BAL) was immediately performed. An enzyme-linked immunosorbent assay was used to measure the expression levels of tumor necrosis factor-α (TNF-α) in the BAL fluid (BALF) and peripheral blood. In addition, the lower lobe of the right lung was removed in order to measure the protein expression levels of intercellular adhesion molecule-1 (ICAM-1) and TNF-α. The results demonstrated that in the rabbits of the TG PaO2 was increased, PaCO2 was decreased, the lung tissue congestion edema was attenuated, the expression levels of TNF-α in the peripheral blood and BALF were reduced and the protein expression levels of ICAM-1 and TNF-α in the lung tissue samples were decreased, as compared with those in the I/RG rabbits. These results suggest that XBJ may protect against left ventricular I/R-induced ALI by regulating the expression of the inflammatory mediators TNF-α and ICAM-1.
Collapse
Affiliation(s)
- Mingli Ji
- Department of Physiology, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Yuxia Wang
- Department of Pathophysiology, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Lei Wang
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Liping Chen
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Jing Li
- Department of Otorhinolaryngology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| |
Collapse
|
86
|
Singh V, Roth S, Veltkamp R, Liesz A. HMGB1 as a Key Mediator of Immune Mechanisms in Ischemic Stroke. Antioxid Redox Signal 2016; 24:635-51. [PMID: 26493086 DOI: 10.1089/ars.2015.6397] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
SIGNIFICANCE Stroke is the leading cause of morbidity and mortality worldwide. Inflammatory cascades have a major impact on outcome and regeneration after ischemic stroke. High-mobility group box 1 (HMGB1) has come into the focus of experimental and clinical stroke research because it is released from necrotic brain tissue and its differential redox forms attract and activate immune cells after ischemic brain injury. HMGB1 is a potent inducer of inflammatory cascades, and thereby, secondary deterioration of neurological outcome. RECENT ADVANCES The role of HMGB1 in sterile inflammation is well established. Emerging evidence suggests that HMGB1 modulates neuroinflammation after experimental brain ischemia and that it may be a useful prognostic biomarker for stroke patients. CRITICAL ISSUES HMGB1 is instantly released from necrotic cells in the ischemic core and activates an early inflammatory response. In addition, brain-released HMGB1 can be redox modified in the circulation and activate peripheral immune cells. HMGB1 concentrations correlate with disease severity and outcome after brain injury. This is the first review depicting the crucial role of HMGB1 in the initiation and perpetuation of secondary immune alterations after experimental and clinical stroke. FUTURE DIRECTIONS HMGB1-dependent signaling pathways are on the verge and have the potential to become a central topic in experimental stroke research. Current and upcoming projects in this field will be paving the way for future translational approaches targeting the center of poststroke inflammation to improve stroke recovery and long-term outcome.
Collapse
Affiliation(s)
- Vikramjeet Singh
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| | - Stefan Roth
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| | - Roland Veltkamp
- 3 Division of Brain Sciences, Imperial College London , London, United Kingdom
| | - Arthur Liesz
- 1 Institute for Stroke and Dementia Research , Klinikum der Universität München, Munich, Germany .,2 Munich Cluster for Systems Neurology (SyNergy) , Munich, Germany
| |
Collapse
|
87
|
Liesz A, Kleinschnitz C. Regulatory T Cells in Post-stroke Immune Homeostasis. Transl Stroke Res 2016; 7:313-21. [DOI: 10.1007/s12975-016-0465-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/15/2016] [Accepted: 03/21/2016] [Indexed: 01/01/2023]
|
88
|
Hocum Stone LL, Xiao F, Rotschafer J, Nan Z, Juliano M, Sanberg CD, Sanberg PR, Kuzmin-Nichols N, Grande A, Cheeran MCJ, Low WC. Amelioration of Ischemic Brain Injury in Rats With Human Umbilical Cord Blood Stem Cells: Mechanisms of Action. Cell Transplant 2016; 25:1473-88. [PMID: 26996530 DOI: 10.3727/096368916x691277] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Despite the high prevalence and devastating outcome, there remain a few options for treatment of ischemic stroke. Currently available treatments are limited by a short time window for treatment and marginal efficacy when used. We have tested a human umbilical cord blood-derived stem cell line that has been shown to result in a significant reduction in stroke infarct volume as well as improved functional recovery following stroke in the rat. In the present study we address the mechanism of action and compared the therapeutic efficacy of high- versus low-passage nonhematopoietic umbilical cord blood stem cells (nh-UCBSCs). Using the middle cerebral arterial occlusion (MCAo) model of stroke in Sprague-Dawley rats, we administered nh-UCBSC by intravenous (IV) injection 2 days following stroke induction. These human cells were injected into rats without any immune suppression, and no adverse reactions were detected. Both behavioral and histological analyses have shown that the administration of these cells reduces the infarct volume by 50% as well as improves the functional outcome of these rats following stroke for both high- and low-passaged nh-UCBSCs. Flow cytometry analysis of immune cells present in the brains of normal rats, rats with ischemic brain injury, and ischemic animals with nh-UCBSC treatment confirmed infiltration of macrophages and T cells consequent to ischemia and reduction to normal levels with nh-UCBSC treatment. Flow cytometry also revealed a restoration of normal levels of microglia in the brain following treatment. These data suggest that nh-UCBSCs may act by inhibiting immune cell migration into the brain from the periphery and possibly by inhibition of immune cell activation within the brain. nh-UCBSCs exhibit great potential for treatment of stroke, including the fact that they are associated with an increased therapeutic time window, no known ill-effects, and that they can be expanded to high numbers for, and stored for, treatment.
Collapse
|
89
|
Systematic review of survival time in experimental mouse stroke with impact on reliability of infarct estimation. J Neurosci Methods 2016; 261:10-8. [PMID: 26620203 DOI: 10.1016/j.jneumeth.2015.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/24/2015] [Accepted: 11/17/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Stroke is the second most common cause of death worldwide. Only one treatment for acute ischemic stroke is currently available, thrombolysis with rt-PA, but it is limited in its use. Many efforts have been invested in order to find additive treatments, without success. A multitude of reasons for the translational problems from mouse experimental stroke to clinical trials probably exists, including infarct size estimations around the peak time of edema formation. Furthermore, edema is a more prominent feature of stroke in mice than in humans, because of the tendency to produce larger infarcts with more substantial edema. PURPOSE This paper will give an overview of previous studies of experimental mouse stroke, and correlate survival time to peak time of edema formation. Furthermore, investigations of whether the included studies corrected the infarct measurements for edema and a comparison of correction methods will be discussed. METHOD Relevant terms were searched in the National Library of Medicine PubMed database. A method for classification of infarct measurement methods was made using a naming convention. CONCLUSION Our study shows that infarct size estimations are often performed around the peak time of edema, with a median of 24h. Most studies do consider edema formation, however, there is no consensus on what method to use to correct for edema. Furthermore, investigations into neuroprotective drugs should use longer survival times to ensure completion of the investigated process. Our findings indicate a need for more research in this area, and establishment of common correction methodology.
Collapse
|
90
|
Estrogen induces multiple regulatory B cell subtypes and promotes M2 microglia and neuroprotection during experimental autoimmune encephalomyelitis. J Neuroimmunol 2016; 293:45-53. [PMID: 27049561 DOI: 10.1016/j.jneuroim.2016.02.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 02/10/2016] [Accepted: 02/11/2016] [Indexed: 01/06/2023]
Abstract
Sex hormones promote immunoregulatory effects on multiple sclerosis. The current study evaluated estrogen effects on regulatory B cells and resident CNS microglia during experimental autoimmune encephalomyelitis (EAE). Herein, we demonstrate an estrogen-dependent induction of multiple regulatory B cell markers indicative of IL-10 dependent as well as IFN-γ dependent pathways. Moreover, although estrogen pretreatment of EAE mice inhibited the infiltration of pro-inflammatory cells into the CNS, it enhanced the frequency of regulatory B cells and M2 microglia. Our study suggests that estrogen has a broad effect on the development of regulatory B cells during EAE, which in turn could promote neuroprotection.
Collapse
|
91
|
Gesuete R, Stevens SL, Stenzel-Poore MP. Role of Circulating Immune Cells in Stroke and Preconditioning-Induced Protection. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:39-44. [PMID: 26463920 DOI: 10.1007/978-3-319-18497-5_7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stroke activates an inflammatory response that results in the infiltration of peripheral immune cells into the ischemic area, contributing to exacerbation of tissue damage. However, evidence indicates that inflammatory cell infiltration can also promote neuroprotection through regulatory immune cells that mitigate injury. These immune regulatory cells may also be important mediators of neuroprotection associated with preconditioning, a phenomenon whereby small exposure to a potential harmful stimulus is able to induce protection against a subsequent ischemic event. The elucidation of mechanisms that allow these immune cells to confer neuroprotection is critical to developing new therapeutic strategies against acute stroke. In the present review, we discuss the dual role of peripheral immune cells in stroke-related brain injury and neuroprotection. Furthermore, we report new data from our laboratory that supports the important role of peripheral cells and their interaction with the brain endothelium for the establishment of the protective phenotype in preconditioning.
Collapse
Affiliation(s)
- Raffaella Gesuete
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Susan L Stevens
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Mary P Stenzel-Poore
- Department of Molecular Microbiology and Immunology, L220, Oregon Health and Science University, 3181 Sam Jackson Park Road, Portland, OR, 97239, USA.
| |
Collapse
|
92
|
Kurnellas MP, Ghosn EEB, Schartner JM, Baker J, Rothbard JJ, Negrin RS, Herzenberg LA, Fathman CG, Steinman L, Rothbard JB. Amyloid fibrils activate B-1a lymphocytes to ameliorate inflammatory brain disease. Proc Natl Acad Sci U S A 2015; 112:15016-23. [PMID: 26621719 PMCID: PMC4679000 DOI: 10.1073/pnas.1521206112] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Amyloid fibrils composed of peptides as short as six amino acids are therapeutic in experimental autoimmune encephalomyelitis (EAE), reducing paralysis and inflammation, while inducing several pathways of immune suppression. Intraperitoneal injection of fibrils selectively activates B-1a lymphocytes and two populations of resident macrophages (MΦs), increasing IL-10 production, and triggering their exodus from the peritoneum. The importance of IL-10-producing B-1a cells in this effective therapy was established in loss-of-function experiments where neither B-cell-deficient (μMT) nor IL10(-/-) mice with EAE responded to the fibrils. In gain-of-function experiments, B-1a cells, adoptively transferred to μMT mice with EAE, restored their therapeutic efficacy when Amylin 28-33 was administered. Stimulation of adoptively transferred bioluminescent MΦs and B-1a cells by amyloid fibrils resulted in rapid (within 60 min of injection) trafficking of both cell types to draining lymph nodes. Analysis of gene expression indicated that the fibrils activated the CD40/B-cell receptor pathway in B-1a cells and induced a set of immune-suppressive cell-surface proteins, including BTLA, IRF4, and Siglec G. Collectively, these data indicate that the fibrils activate B-1a cells and F4/80(+) MΦs, resulting in their migration to the lymph nodes, where IL-10 and cell-surface receptors associated with immune-suppression limit antigen presentation and T-cell activation. These mechanisms culminate in reduction of paralytic signs of EAE.
Collapse
Affiliation(s)
- Michael Phillip Kurnellas
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Eliver Eid Bou Ghosn
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - Jill M Schartner
- Division of Immunology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jeanette Baker
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jesse J Rothbard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305
| | - Robert S Negrin
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Leonore A Herzenberg
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - C Garrison Fathman
- Division of Immunology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Lawrence Steinman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305;
| | - Jonathan B Rothbard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305; Division of Immunology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
93
|
Partial MHC Constructs Treat Thromboembolic Ischemic Stroke Characterized by Early Immune Expansion. Transl Stroke Res 2015; 7:70-8. [PMID: 26627498 DOI: 10.1007/s12975-015-0436-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/19/2015] [Accepted: 11/20/2015] [Indexed: 12/22/2022]
Abstract
Inflammation and thrombosis are tightly linked, with inflammation contributing to thromboembolism and to stroke outcome. Thromboembolism is a frequent cause of ischemic stroke; yet, the most used occlusion mouse models of experimental stroke do not effectively replicate thromboembolism. Our group recently described a novel thromboembolic mouse model of stroke that successfully occludes the middle cerebral artery with high reproducibility. In the current study, we characterize the peripheral and local immune outcomes as well as the ischemic response to immune therapy in a clinically relevant mouse model of thromboembolic stroke. Brain and spleen tissues were harvested 24 h after thromboembolic stroke and cells immunophenotyped by flow cytometry. We observed a significant increase in neutrophils and early activated T cells in the spleen and an increase in neutrophils and activated monocytes/microglia in the ischemic cortex after thromboembolic stroke. Moreover, as was shown previously for transient MCAO models, treatment of thromboembolic stroke with partial MHC constructs significantly reduced ischemic damage indicating an equivalent effect of this immune-based therapy in the thromboembolic model that better mimics the pathophysiology of human stroke.
Collapse
|
94
|
Bodhankar S, Lapato A, Chen Y, Vandenbark AA, Saugstad JA, Offner H. Role for microglia in sex differences after ischemic stroke: importance of M2. Metab Brain Dis 2015; 30:1515-29. [PMID: 26246072 PMCID: PMC4644102 DOI: 10.1007/s11011-015-9714-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 07/20/2015] [Indexed: 01/17/2023]
Abstract
Inflammation plays a critical role in the pathogenesis of ischemic stroke. This process depends, in part, upon proinflammatory factors released by activated resident central nervous system (CNS) microglia (MG). Previous studies demonstrated that transfer of IL-10(+) B-cells reduced infarct volumes in male C57BL/6 J recipient mice when given 24 h prior to or therapeutically at 4 or 24 h after experimental stroke induced by 60 min middle cerebral artery occlusion (MCAO). The present study assesses possible sex differences in immunoregulation by IL-10(+) B-cells on primary male vs. female MG cultured from naïve and ischemic stroke-induced mice. Thus, MG cultures were treated with recombinant (r)IL-10, rIL-4 or IL-10(+) B-cells after lipopolysaccharide (LPS) activation and evaluated by flow cytometry for production of proinflammatory and anti-inflammatory factors. We found that IL-10(+) B-cells significantly reduced MG production of TNF-α, IL-1β and CCL3 post-MCAO and increased their expression of the anti-inflammatory M2 marker, CD206, by cell-cell interactions. Moreover, MG from female vs. male mice had higher expression of IL-4 and IL-10 receptors and increased production of IL-4, especially after treatment with IL-10(+) B-cells. These findings indicate that IL-10-producing B-cells play a crucial role in regulating MG activation, proinflammatory cytokine release and M2 phenotype induction, post-MCAO, with heightened sensitivity of female MG to IL-4 and IL-10. This study, coupled with our previous demonstration of increased numbers of transferred IL-10(+) B-cells in the ischemic hemisphere, provide a mechanistic basis for local regulation by secreted IL-10 and IL-4 as well as direct B-cell/MG interactions that promote M2-MG.
Collapse
Affiliation(s)
- Sheetal Bodhankar
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA
| | - Andrew Lapato
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA
| | - Yingxin Chen
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA
| | - Arthur A Vandenbark
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA
| | - Julie A Saugstad
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA
- Department of Medical and Molecular Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA
| | - Halina Offner
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA.
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA.
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, USA.
| |
Collapse
|
95
|
Zhang J, Lapato A, Bodhankar S, Vandenbark AA, Offner H. Treatment with IL-10 producing B cells in combination with E2 ameliorates EAE severity and decreases CNS inflammation in B cell-deficient mice. Metab Brain Dis 2015; 30:1117-27. [PMID: 25776868 PMCID: PMC4561218 DOI: 10.1007/s11011-015-9661-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 02/17/2015] [Indexed: 11/29/2022]
Abstract
Clinical improvement during pregnancy in multiple sclerosis (MS) patients suggests that sex hormones exert potent regulatory effects on autoimmune function. Our previous studies demonstrated that estrogen- (17β-estradiol; E2) mediated protection against experimental autoimmune encephalomyelitis (EAE), a mouse model for MS, hinges on the B cells, leading to elevated numbers of IL-10 secreting CD1d(hi)CD5(+) B regulatory cells (Bregs) in wild type mice. Our data show that co-administration of E2 and IL-10(+) B cells ameliorates EAE disease severity and limits CNS infiltrating leukocytes in B cell deficient mice. Additionally, treatment with E2 and Bregs reduces demyelination and dramatically decreases the proportion of CD11b(+)CD45(hi) activated microglia/macrophages found in the CNS of immunized animals compared to vehicle, E2 or Breg cells alone. Furthermore, mice given E2 and Bregs exhibit increased numbers of peripheral programmed death-1 positive CD4(+)Foxp3(+) regulatory T cells (Tregs) and up-regulation of programmed death receptor-ligand-1 and CD80 expression on monocytes. Our study suggests IL-10 producing Bregs have powerful therapeutic potential as an agent against EAE when augmented with E2 treatment.
Collapse
Affiliation(s)
- Jun Zhang
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Andrew Lapato
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Sheetal Bodhankar
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
| | - Arthur A. Vandenbark
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR
- Correspondence author: Halina Offner, Neuroimmunology Research R&D-31, VA Portland Health Care System, 3710 SW US Veterans Hospital Rd., Portland, OR 97239, USA.
| | - Halina Offner
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR
| |
Collapse
|
96
|
Liang QJ, Jiang M, Wang XH, Le LL, Xiang M, Sun N, Meng D, Chen SF. Pre-existing interleukin 10 in cerebral arteries attenuates subsequent brain injury caused by ischemia/reperfusion. IUBMB Life 2015; 67:710-9. [PMID: 26337686 DOI: 10.1002/iub.1429] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/20/2015] [Indexed: 01/05/2023]
Abstract
Recurrent stroke is difficult to treat and life threatening. Transfer of anti-inflammatory gene is a potential gene therapy strategy for ischemic stroke. Using recombinant adeno-associated viral vector 1 (rAAV1)-mediated interleukin 10 (IL-10), we investigated whether transfer of beneficial gene into the rat cerebral vessels during interventional treatment for initial stroke could attenuate brain injury caused by recurrent stroke. Male Wistar rats were administered rAAV1-IL-10, rAAV1-YFP, or saline into the left cerebral artery. Three weeks after gene transfer, rats were subjected to occlusion of the left middle cerebral artery (MCAO) for 45 min followed by reperfusion for 24 h. IL-10 levels in serum were significantly elevated 3 weeks after rAAV1-IL-10 injection, and virus in the cerebral vessels was confirmed by in situ hybridization. Pre-existing IL-10 but not YFP decreased the neurological dysfunction scores, brain infarction volume, and the number of injured neuronal cells. AAV1-IL-10 transduction increased heme oxygenase (HO-1) mRNA and protein levels in the infarct boundary zone of the brain. Thus, transduction of the IL-10 gene in the cerebral artery prior to ischemia attenuates brain injury caused by ischemia/reperfusion in rats. This preventive approach for recurrent stroke can be achieved during interventional treatment for initial stroke.
Collapse
Affiliation(s)
- Qiu-Juan Liang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai, 200032, China
| | - Mei Jiang
- Department of Neurology, Gongli Hospital, Shanghai, 200135, China
| | - Xin-Hong Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai, 200032, China
| | - Li-Li Le
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai, 200032, China
| | - Meng Xiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai, 200032, China
| | - Ning Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai, 200032, China
| | - Dan Meng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai, 200032, China
| | - Si-Feng Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai, 200032, China
| |
Collapse
|
97
|
Bodhankar S, Chen Y, Lapato A, Dotson AL, Wang J, Vandenbark AA, Saugstad JA, Offner H. PD-L1 Monoclonal Antibody Treats Ischemic Stroke by Controlling Central Nervous System Inflammation. Stroke 2015; 46:2926-34. [PMID: 26306753 DOI: 10.1161/strokeaha.115.010592] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/05/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Both pathogenic and regulatory immune processes are involved in the middle cerebral artery occlusion (MCAO) model of experimental stroke, including interactions involving the programmed death 1 (PD-1) receptor and its 2 ligands, PD-L1 and PD-L2. Although PD-1 reduced stroke severity, PD-L1 and PD-L2 appeared to play pathogenic roles, suggesting the use of anti-PD-L monoclonal antibody therapy for MCAO. METHODS Male C57BL/6 mice were treated with a single dose of anti-PD-L1 monoclonal antibody 4 hours after MCAO and evaluated for clinical, histological and immunologic changes after 96 hours of reperfusion. RESULTS Blockade of the PD-L1 checkpoint using a single injection of 200 μg anti-PD-L1 monoclonal antibody given intravenously 4 hours after occlusion significantly reduced MCAO infarct volumes and improved neurological outcomes after 96 hours of reperfusion. Treatment partially reversed splenic atrophy and decreased central nervous system infiltrating immune cells concomitant with enhanced appearance of CD8(+) regulatory T cells in the lesioned central nervous system hemisphere. CONCLUSIONS This study demonstrates for the first time the beneficial therapeutic effects of PD-L1 checkpoint blockade on MCAO, thus validating proposed mechanisms obtained in our previous studies using PD-1- and PD-L-deficient mice. These results provide strong support for the use of available humanized anti-PD-L1 antibodies for treatment of human stroke subjects.
Collapse
Affiliation(s)
- Sheetal Bodhankar
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Yingxin Chen
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Andrew Lapato
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Abby L Dotson
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Jianming Wang
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Arthur A Vandenbark
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Julie A Saugstad
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland
| | - Halina Offner
- From the Neuroimmunology Research, VA Portland Health Care System, OR (S.B., A.L., A.L.D., A.A.V., H.O.); and Departments of Neurology (S.B., A.L., A.L.D., A.A.V., J.A.S., H.O.), Anesthesiology and Perioperative Medicine (Y.C., J.W., J.A.S., H.O.), Molecular Microbiology and Immunology (A.A.V.), and Medical and Molecular Genetics (J.A.S.), Oregon Health and Science University, Portland.
| |
Collapse
|
98
|
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: 44] [Impact Index Per Article: 4.9] [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
|
99
|
|
100
|
Zhang J, Benedek G, Bodhankar S, Lapato A, Vandenbark AA, Offner H. IL-10 producing B cells partially restore E2-mediated protection against EAE in PD-L1 deficient mice. J Neuroimmunol 2015. [PMID: 26198929 DOI: 10.1016/j.jneuroim.2015.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Women with multiple sclerosis (MS) often experience clinical improvement during pregnancy, indicating that sex hormones might have therapeutic effects in MS. Our previous studies have demonstrated that B cells and PD-L1 are crucial for E2 (17β-estradiol)-mediated protection against experimental autoimmune encephalomyelitis (EAE). We here demonstrate that the transfer of IL-10(+) B cells into E2-treated PD-L1(-/-) mice after EAE induction could partially restore E2-mediated protection and decrease the frequency of pro-inflammatory cells in the CNS compared to E2/saline treated PD-L1(-/-) mice. Hence, co-administration of IL-10(+) B cells and E2 might have a powerful therapeutic potential for treatment of EAE.
Collapse
Affiliation(s)
- Jun Zhang
- 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.
| | - Sheetal Bodhankar
- Neuroimmunology Research, VA Portland Health Care System, Portland, OR, USA; Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
| | - Andrew Lapato
- 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.
| | - 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 and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|