The immunology of stroke: from mechanisms to translation

Costantino Iadecola, Josef Anrather, Costantino Iadecola, Josef Anrather

Abstract

Immunity and inflammation are key elements of the pathobiology of stroke, a devastating illness second only to cardiac ischemia as a cause of death worldwide. The immune system participates in the brain damage produced by ischemia, and the damaged brain, in turn, exerts an immunosuppressive effect that promotes fatal infections that threaten the survival of people after stroke. Inflammatory signaling is involved in all stages of the ischemic cascade, from the early damaging events triggered by arterial occlusion to the late regenerative processes underlying post-ischemic tissue repair. Recent developments have revealed that stroke engages both innate and adaptive immunity. But adaptive immunity triggered by newly exposed brain antigens does not have an impact on the acute phase of the damage. Nevertheless, modulation of adaptive immunity exerts a remarkable protective effect on the ischemic brain and offers the prospect of new stroke therapies. As immunomodulation is not devoid of deleterious side effects, a better understanding of the reciprocal interaction between the immune system and the ischemic brain is essential to harness the full therapeutic potential of the immunology of stroke.

Figures

Figure 1. Early vascular, perivascular and parenchymal…
Figure 1. Early vascular, perivascular and parenchymal events triggered by I/R
Hypoxia, ROS and changes in shear stress initiate the cellular events induced by I/R. In the vessels lumen, I/R leads to blood clotting, platelet aggregation and cytokine release (IL-1α). Translocation of P-selectin on the surface of platelets and EC leads to platelet-leukocyte aggregation. Complement is activated and arachidonic acid metabolites are released. In the vascular wall, upregulation of E- and P-selectin on EC provides a platform for low affinity leukocyte binding through interaction with sialyl Lex moieties of glycoproteins expressed on leukocytes, e.g. PSGL-1. Firm adhesion is obtained after endothelial expression of ICAM-1 interacting with leukocyte β2 integrins (LFA-1 and Mac-1). Loss of NO promotes vasoconstriction and enhances leukocyte and platelet aggregation. MMP activation could lead to BBB breakdown and matrix proteolysis facilitating leukocyte extravasation. In the perivascular space, chemotactic complement subunits (C5a) acting on mast cell complement receptors (CD88) leads to degranulation and release of histamine and proteases, contributing to BBB leakiness. Cytokines (TNF, IL-1β) are produced by mast cells and perivascular macrophages, providing further signals to guide leukocyte migration across the vessel wall,. In the brain parenchyma, injured cells release purines (ATP), which act as early proinflammatory signals leading to production of cytokines and chemokines. Disruption of neuronal-microglial interaction (CX3CL1, CD200) and increases in extracellular glutamate (glu) acting on microglial GluR1 metabotropic receptor also contribute to the pro-inflammatory milieu.
Figure 2. Cell death and activation of…
Figure 2. Cell death and activation of pattern recognition receptors set the stage adaptive immunity
Release of nucleotides (ATP, UTP) from injured cells, including neurons, activates purinergic receptors on microglia and macrophages, and leads to production of pro-inflammatory cytokines. While most of these cytokines are transcriptionally induced, IL-1β and IL-18 are processed from their pro-peptides by the activity of interleukin-1 converting enzyme (ICE; caspase 1). ICE is embedded in a multi-protein complex (NLRP3 or inflammasome) and is activated by microglial P2×7 receptors. Ischemic cell death leads to the formation of danger associated molecular patterns (DAMPs) molecules, which activate TLRs, especially TLR2 and 4. DAMPs released by ischemia include HMGB1, an intracellular DNA binding protein released after cellular injury, HSP60, and β-amyloid (Aβ), among others. TLRs, in concert with scavenger receptors such as CD36, upregulate pro-inflammatory gene expression through the transcription factor NF-κB,. DAMPs also derive from matrix breakdown by lytic enzymes released from dead cells and by the action of reactive oxygen species (ROS) on lipids. The cytokine production and complement activation resulting from these events leads to increased leukocyte infiltration and enhances tissue damage, which, in turn, produces more DAMPs. Antigens unveiled by tissue damage are presented to T cells, setting the stage for adaptive immunity.
Figure 3. Deleterious and beneficial roles of…
Figure 3. Deleterious and beneficial roles of T cells in stroke
In the acute phase of cerebral ischemia, unprimed T cells contribute to tissue damage in an antigen independent manner (innate immunity), possibly through IFNγ and ROS (left upper quadrant). γδT cells, activated by IL-23 released from microglia/macrophages, produce the cytotoxic cytokine IL-17 and contributes to acute ischemic brain injury. However, T cells can also be protective. TGFβ produced by neurons, glia, or macroglia/macrophages promotes the development of Treg cells secreting the protective cytokine IL-10 and inhibits Th1 and Th2 responses. Treg cells are protective in models of cerebral ischemia. Induction of mucosal tolerance with CNS antigens produces an adaptive response, which leads to the establishment of autoreactive Th2 cells producing IL-10 and Treg cells producing IL-10 and TGFβ, is highly protective in experimental stroke (right lower quadrant). As discussed in the text, there is no evidence that adaptive immunity contributes to acute ischemic brain injury. However, weeks and months after stroke, autoreactive CD4+ and CD8+ T cells targeting CNS antigens could develop (right upper quadrant). The resulting cell death could play a role in the delayed brain damage and atrophy that occurs after stroke.
Figure 4. Resolution of inflammation and tissue…
Figure 4. Resolution of inflammation and tissue repair
Clearing of dead cells and suppression of inflammation are key events in brain repair. “Find-me signals” (UTP, ATP) attract microglia and macrophages through P2Y2 receptors. “Eat-me signals” include UDP, which act on P2Y6 receptors to stimulate microglial phagocytosis, and phosphatidylserine (PtdSer), which is translocated to the outer leaflet of the plasma membrane of apoptotic cells. PtdSer binding proteins involved in the clearance of dead cells include MGF-E8 on microglia and TIM4 on macrophages. Immunoglobulins directed against CNS antigens, which appear after stroke (Table 2), may also promote phagocytosis by engaging Fc receptors on phagocytic cells. Phagocytosis promotes secretion of IL-10 and TGFβ, which, in turn, suppress antigen presentation, promote Treg formation, inhibit expression of adhesion molecules in EC and production of proinflammatory cytokines,. TGFβ and IL-10 are also neuroprotective, and may facilitate brain repair processes. In addition, arachidonic and omega-3 fatty acids metabolites lipoxins, resolvins, and protectins, which play an active role in the resolution of inflammation in other organs, could also contribute to suppress post-ischemic inflammation. Growth factors and MMPs produced by EC, neurons, astrocytes, oligodendrocytes and microglia are critical molecules driving tissue reorganization and repair,.

References

    1. Mena H, Cadavid D, Rushing EJ. Human cerebral infarct: a proposed histopathologic classification based on 137 cases. Acta Neuropathol. 2004;108:524–530.
    1. Moskowitz MA, Lo EH, Iadecola C. The science of stroke: mechanisms in search of treatments. Neuron. 2010;67:181–198.
    1. Meisel C, Schwab J, Prass K, Meisel A, Dirnagl U. Central nervous system injury-induced immune deficiency syndrome. Nat Rev Neurosci. 2005;6:775–786.
    1. Urra X, Cervera A, Villamor N, Planas AM, Chamorro A. Harms and benefits of lymphocyte subpopulations in patients with acute stroke. Neuroscience. 2009;158:1174–1183.
    1. Weiner HL. The challenge of multiple sclerosis: how do we cure a chronic heterogeneous disease? Ann Neurol. 2009;65:239–248.
    1. Carden DL, Granger DN. Pathophysiology of ischaemia-reperfusion injury. J Pathol. 2000;190:255–266.
    1. Peerschke EI, Yin W, Ghebrehiwet B. Complement activation on platelets: implications for vascular inflammation and thrombosis. Mol Immunol. 2010;47:2170–2175.
    1. Pinsky DJ, et al. Hypoxia-induced exocytosis of endothelial cell Weibel-Palade bodies. A mechanism for rapid neutrophil recruitment after cardiac preservation. The Journal of clinical investigation. 1996;97:493–500.
    1. Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J Med. 2011;364:656–665.
    1. Del Zoppo GJ, Schmid-Schonbein GW, Mori E, Copeland BR, Chang CM. Polymorphonuclear leukocytes occlude capillaries following middle cerebral artery occlusion and reperfusion in baboons. Stroke. 1991;22:1276–1283.
    1. Hyman MC, et al. Self-regulation of inflammatory cell trafficking in mice by the leukocyte surface apyrase CD39. The Journal of clinical investigation. 2009;119:1136–1149.
    1. Yilmaz G, Granger DN. Leukocyte recruitment and ischemic brain injury. Neuromolecular Med. 2010;12:193–204.
    1. Atochin DN, et al. The phosphorylation state of eNOS modulates vascular reactivity and outcome of cerebral ischemia in vivo. The Journal of clinical investigation. 2007;117:1961–1967.
    1. Ishikawa M, Zhang JH, Nanda A, Granger DN. Inflammatory responses to ischemia and reperfusion in the cerebral microcirculation. Front Biosci. 2004;9:1339–1347.
    1. Yemisci M, et al. Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat Med. 2009;15:1031–1037.
    1. Engelhardt B, Sorokin L. The blood-brain and the blood-cerebrospinal fluid barriers: function and dysfunction. Semin Immunopathol. 2009;31:497–511.
    1. Lindsberg PJ, Strbian D, Karjalainen-Lindsberg ML. Mast cells as early responders in the regulation of acute blood-brain barrier changes after cerebral ischemia and hemorrhage. J Cereb Blood Flow Metab. 2010;30:689–702.
    1. Strbian D, Karjalainen-Lindsberg ML, Tatlisumak T, Lindsberg PJ. Cerebral mast cells regulate early ischemic brain swelling and neutrophil accumulation. J Cereb Blood Flow Metab. 2006;26:605–612.
    1. Konsman JP, Drukarch B, Van Dam AM. (Peri)vascular production and action of pro-inflammatory cytokines in brain pathology. Clin Sci (Lond) 2007;112:1–25.
    1. Sairanen TR, Lindsberg PJ, Brenner M, Siren AL. Global forebrain ischemia results in differential cellular expression of interleukin-1beta (IL-1beta) and its receptor at mRNA and protein level. J Cereb Blood Flow Metab. 1997;17:1107–1120.
    1. Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol. 2008;8:279–289.
    1. Bours MJ, Swennen EL, Di Virgilio F, Cronstein BN, Dagnelie PC. Adenosine 5'-triphosphate and adenosine as endogenous signaling molecules in immunity and inflammation. Pharmacol Ther. 2006;112:358–404.
    1. Melani A, et al. ATP extracellular concentrations are increased in the rat striatum during in vivo ischemia. Neurochem Int. 2005;47:442–448.
    1. Schock SC, et al. Cortical spreading depression releases ATP into the extracellular space and purinergic receptor activation contributes to the induction of ischemic tolerance. Brain Res. 2007;1168:129–138.
    1. Burnstock G. Purinergic signalling and disorders of the central nervous system. Nat Rev Drug Discov. 2008;7:575–590.
    1. Bune LT, Thaning P, Johansson PI, Bochsen L, Rosenmeier JB. Effects of nucleotides and nucleosides on coagulation. Blood Coagul Fibrinolysis. 2010;21:436–441.
    1. Pocock JM, Kettenmann H. Neurotransmitter receptors on microglia. Trends Neurosci. 2007;30:527–535.
    1. Peachell P. Regulation of mast cells by β-agonists. Clin Rev Allergy Immunol. 2006;31:131–142.
    1. Samson MT, et al. Differential roles of CB1 and CB2 cannabinoid receptors in mast cells. J Immunol. 2003;170:4953–4962.
    1. Marsh BJ, Williams-Karnesky RL, Stenzel-Poore MP. Toll-like receptor signaling in endogenous neuroprotection and stroke. Neuroscience. 2009;158:1007–1020.
    1. Hoek RM, et al. Down-regulation of the macrophage lineage through interaction with OX2 (CD200) Science. 2000;290:1768–1771.
    1. Matsumoto H, et al. Expression of CD200 by macrophage-like cells in ischemic core of rat brain after transient middle cerebral artery occlusion. Neurosci Lett. 2007;418:44–48.
    1. Cardona AE, et al. Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006;9:917–924.
    1. Chapman GA, et al. Fractalkine cleavage from neuronal membranes represents an acute event in the inflammatory response to excitotoxic brain damage. J Neurosci. 2000;20:RC87.
    1. Felger JC, et al. Brain dendritic cells in ischemic stroke: time course, activation state, and origin. Brain Behav Immun. 2010;24:724–737.
    1. Kostulas N, et al. Dendritic cells are present in ischemic brain after permanent middle cerebral artery occlusion in the rat. Stroke. 2002;33:1129–1134.
    1. Gelderblom M, et al. Temporal and spatial dynamics of cerebral immune cell accumulation in stroke. Stroke. 2009;40:1849–1857.
    1. Yilmaz A, et al. Transient decrease in circulating dendritic cell precursors after acute stroke: potential recruitment into the brain. Clin Sci. 2010;118:147–157.
    1. Hurn PD, et al. T- and B-cell-deficient mice with experimental stroke have reduced lesion size and inflammation. J Cereb Blood Flow Metab. 2007;27:1798–1805.
    1. Kleinschnitz C, et al. Early detrimental T-cell effects in experimental cerebral ischemia are neither related to adaptive immunity nor thrombus formation. Blood. 2010;115:3835–3842.
    1. Shichita T, et al. Pivotal role of cerebral interleukin-17-producing γδT cells in the delayed phase of ischemic brain injury. Nat Med. 2009;15:946–950.
    1. Liesz A, et al. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat Med. 2009;15:192–199.
    1. Ren X, Akiyoshi K, Vandenbark AA, Hurn PD, Offner H. CD4(+)FoxP3(+) regulatory T-cells in cerebral ischemic stroke. Metab Brain Dis. 2010
    1. Becker KJ, et al. Immunologic tolerance to myelin basic protein decreases stroke size after transient focal cerebral ischemia. Proc Natl Acad Sci U S A. 1997;94:10873–10878.
    1. Frenkel D, et al. Nasal vaccination with myelin oligodendrocyte glycoprotein reduces stroke size by inducing IL-10-producing CD4+ T cells. Journal of immunology (Baltimore, Md : 1950) 2003;171:6549–6555.
    1. Becker K, Kindrick D, McCarron R, Hallenbeck J, Winn R. Adoptive transfer of myelin basic protein-tolerized splenocytes to naive animals reduces infarct size: a role for lymphocytes in ischemic brain injury? Stroke. 2003;34:1809–1815.
    1. Becker KJ. Sensitization and tolerization to brain antigens in stroke. Neuroscience. 2009;158:1090–1097.
    1. Frenkel D, et al. Neuroprotection by IL-10-producing MOG CD4+ T cells following ischemic stroke. J Neurol Sci. 2005;233:125–132.
    1. Subramanian S, et al. Recombinant T cell receptor ligand treats experimental stroke. Stroke. 2009;40:2539–2545.
    1. Itakura A, et al. Characterization of human platelet binding of recombinant T cell receptor ligand. Journal of neuroinflammation. 2010;7:75.
    1. Abbas Ak. Basic Immunology Updated Edition: Functions and Disorders of the Immune System. Saunders; 2010.
    1. Yilmaz G, Arumugam TV, Stokes KY, Granger DN. Role of T lymphocytes and interferon-γ in ischemic stroke. Circulation. 2006;113:2105–2112.
    1. Offner H, Vandenbark AA, Hurn PD. Effect of experimental stroke on peripheral immunity: CNS ischemia induces profound immunosuppression. Neuroscience. 2009;158:1098–1111.
    1. Andrews DM, Smyth MJ. A potential role for RAG-1 in NK cell development revealed by analysis of NK cells during ontogeny. Immunol Cell Biol. 2010;88:107–116.
    1. Spite M, Serhan CN. Novel lipid mediators promote resolution of acute inflammation: impact of aspirin and statins. Circ Res. 2010;107:1170–1184.
    1. Nathan C, Ding A. Nonresolving inflammation. Cell. 2010;140:871–882.
    1. Schilling M, et al. Predominant phagocytic activity of resident microglia over hematogenous macrophages following transient focal cerebral ischemia: an investigation using green fluorescent protein transgenic bone marrow chimeric mice. Exp Neurol. 2005;196:290–297.
    1. Denes A, et al. Proliferating resident microglia after focal cerebral ischaemia in mice. J Cereb Blood Flow Metab. 2007;27:1941–1953.
    1. Rappert A, et al. CXCR3-dependent microglial recruitment is essential for dendrite loss after brain lesion. J Neurosci. 2004;24:8500–8509.
    1. Davalos D, et al. ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005;8:752–758.
    1. Taylor A, Verhagen J, Blaser K, Akdis M, Akdis CA. Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: the role of T regulatory cells. Immunology. 2006;117:433–442.
    1. Greenberg DA, Jin K. Growth factors and stroke. NeuroRx. 2006;3:458–465.
    1. Carmichael ST. Translating the frontiers of brain repair to treatments: starting not to break the rules. Neurobiol Dis. 2010;37:237–242.
    1. Zhang ZG, Zhang L, Jiang Q, Chopp M. Bone marrow-derived endothelial progenitor cells participate in cerebral neovascularization after focal cerebral ischemia in the adult mouse. Circ Res. 2002;90:284–288.
    1. Hayakawa K, Qiu J, Lo EH. Biphasic actions of HMGB1 signaling in inflammation and recovery after stroke. Ann N Y Acad Sci. 2010;1207:50–57.
    1. Li S, et al. An age-related sprouting transcriptome provides molecular control of axonal sprouting after stroke. Nat Neurosci. 2010;13:1496–1504.
    1. Lalancette-Hebert M, Gowing G, Simard A, Weng YC, Kriz J. Selective ablation of proliferating microglial cells exacerbates ischemic injury in the brain. J Neurosci. 2007;27:2596–2605.
    1. Hayakawa K, et al. Inhibition of reactive astrocytes with fluorocitrate retards neurovascular remodeling and recovery after focal cerebral ischemia in mice. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2010;30:871–882.
    1. Zhang ZG, et al. Correlation of VEGF and angiopoietin expression with disruption of blood-brain barrier and angiogenesis after focal cerebral ischemia. J Cereb Blood Flow Metab. 2002;22:379–392.
    1. Hao Q, et al. Neutrophil depletion decreases VEGF-induced focal angiogenesis in the mature mouse brain. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2007;27:1853–1860.
    1. Manoonkitiwongsa PS, Schultz RL, McCreery DB, Whitter EF, Lyden PD. Neuroprotection of ischemic brain by vascular endothelial growth factor is critically dependent on proper dosage and may be compromised by angiogenesis. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2004;24:693–702.
    1. Zhang ZG, et al. VEGF enhances angiogenesis and promotes blood-brain barrier leakage in the ischemic brain. The Journal of clinical investigation. 2000;106:829–838.
    1. Denes A, et al. Experimental stroke-induced changes in the bone marrow reveal complex regulation of leukocyte responses. J Cereb Blood Flow Metab. 2010
    1. Jickling GC, et al. Signatures of cardioembolic and large-vessel ischemic stroke. Ann Neurol. 2010;68:681–692.
    1. Offner H, et al. Experimental stroke induces massive, rapid activation of the peripheral immune system. J Cereb Blood Flow Metab. 2006;26:654–665.
    1. Emsley HCA, et al. An early and sustained peripheral inflammatory response in acute ischaemic stroke: relationships with infection and atherosclerosis. J Neuroimmunol. 2003;139:93–101.
    1. Smith CJ, et al. Peak plasma interleukin-6 and other peripheral markers of inflammation in the first week of ischaemic stroke correlate with brain infarct volume, stroke severity and long-term outcome. BMC Neurol. 2004;4:2.
    1. Haeusler KG, et al. Immune responses after acute ischemic stroke or myocardial infarction. International journal of cardiology. 2010
    1. Gendron A, et al. Temporal effects of left versus right middle cerebral artery occlusion on spleen lymphocyte subsets and mitogenic response in Wistar rats. Brain Res. 2002;955:85–97.
    1. Vogelgesang A, et al. Functional status of peripheral blood T-cells in ischemic stroke patients. PLoS ONE. 2010;5:e8718.
    1. Chamorro A, Urra X, Planas AM. Infection after acute ischemic stroke: a manifestation of brain-induced immunodepression. Stroke. 2007;38:1097–1103.
    1. Prass K, et al. Stroke-induced immunodeficiency promotes spontaneous bacterial infections and is mediated by sympathetic activation reversal by poststroke T helper cell type 1-like immunostimulation. J Exp Med. 2003;198:725–736.
    1. Leys D, Henon H, Mackowiak-Cordoliani MA, Pasquier F. Poststroke dementia. Lancet Neurol. 2005;4:752–759.
    1. McColl BW, Rothwell NJ, Allan SM. Systemic inflammatory stimulus potentiates the acute phase and CXC chemokine responses to experimental stroke and exacerbates brain damage via interleukin-1- and neutrophil-dependent mechanisms. J Neurosci. 2007;27:4403–4412.
    1. Becker KJ, Kindrick DL, Lester MP, Shea C, Ye ZC. Sensitization to brain antigens after stroke is augmented by lipopolysaccharide. J Cereb Blood Flow Metab. 2005;25:1634–1644.
    1. Zierath D, et al. CNS immune responses following experimental stroke. Neurocrit Care. 2010;12:274–284.
    1. Fonarow GC, et al. Timeliness of Tissue-Type Plasminogen Activator Therapy in Acute Ischemic Stroke: Patient Characteristics, Hospital Factors, and Outcomes Associated With Door-to-Needle Times Within 60 Minutes. Circulation. 2011;123:750–758.
    1. Gesuete R, et al. Recombinant C1 inhibitor in brain ischemic injury. Ann Neurol. 2009;66:332–342.
    1. Iadecola C, Zhang F, Xu X. Inhibition of inducible nitric oxide synthase ameliorates cerebral ischemic damage. Am J. Physiol. 1995;268:R286–R292.
    1. Lee S-T, et al. Anti-inflammatory mechanism of intravascular neural stem cell transplantation in haemorrhagic stroke. Brain : a journal of neurology. 2008;131:616–629.
    1. Prestigiacomo CJ, et al. CD18-mediated neutrophil recruitment contributes to the pathogenesis of reperfused but not nonreperfused stroke. Stroke. 1999;30:1110–1117.
    1. Stowe AM, et al. Neutrophil elastase and neurovascular injury following focal stroke and reperfusion. Neurobiol Dis. 2009;35:82–90.
    1. del Zoppo GJ. Lessons from stroke trials using anti-inflammatory approaches that have failed. In: Dirnagl U, Elger B, editors. Neuroinflammation in stroke. Berlin - Heidelberg: Springer-Verlagh; 2004. pp. 155–184.
    1. Furuya K, et al. Examination of several potential mechanisms for the negative outcome in a clinical stroke trial of enlimomab, a murine anti-human intercellular adhesion molecule-1 antibody: a bedside-to-bench study. Stroke. 2001;32:2665–2674.
    1. Fagan SC, et al. Minocycline to improve neurologic outcome in stroke (MINOS): a dose-finding study. Stroke; a journal of cerebral circulation. 2010;41:2283–2287.
    1. Jensen HA, et al. Remote ischemic preconditioning protects the brain against injury after hypothermic circulatory arrest. Circulation. 2011;123:714–721.
    1. Kariko K, Weissman D, Welsh FA. Inhibition of toll-like receptor and cytokine signaling--a unifying theme in ischemic tolerance. J Cereb Blood Flow Metab. 2004;24:1288–1304.
    1. Marsh B, et al. Systemic lipopolysaccharide protects the brain from ischemic injury by reprogramming the response of the brain to stroke: a critical role for IRF3. J Neurosci. 2009;29:9839–9849.
    1. Blondeau N, Widmann C, Lazdunski M, Heurteaux C. Activation of the nuclear factor-κB is a key event in brain tolerance. J Neurosci. 2001;21:4668–4677.
    1. Kunz A, et al. Neurovascular protection by ischemic tolerance: role of nitric oxide and reactive oxygen species. J Neurosci. 2007;27:7083–7093.
    1. Ohtsuki T, Ruetzler CA, Tasaki K, Hallenbeck JM. Interleukin-1 mediates induction of tolerance to global ischemia in gerbil hippocampal CA1 neurons. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 1996;16:1137–1142.
    1. Pradillo JM, et al. TNFR1 upregulation mediates tolerance after brain ischemic preconditioning. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism. 2005;25:193–203.
    1. Ziv Y, et al. A novel immune-based therapy for stroke induces neuroprotection and supports neurogenesis. Stroke. 2007;38:774–782.
    1. Steinman L. A brief history of T(H)17, the first major revision in the T(H)1/T(H)2 hypothesis of T cell-mediated tissue damage. Nat Med. 2007;13:139–145.
    1. Schwartz M, London A, Shechter R. Boosting T-cell immunity as a therapeutic approach for neurodegenerative conditions: the role of innate immunity. Neuroscience. 2009;158:1133–1142.
    1. Genain CP, et al. Late complications of immune deviation therapy in a nonhuman primate. Science (New York, NY) 1996;274:2054–2057.
    1. Gee JM, et al. Long term immunologic consequences of experimental stroke and mucosal tolerance. Exp Transl Stroke Med. 2009;1:3.
    1. Lipton P. Ischemic Cell Death in Brain Neurons. Physiol Rev. 1999;79:1431–1568.
    1. Bacigaluppi M, Comi G, Hermann DM. Animal models of ischemic stroke. Part one: modeling risk factors. Open Neurol J. 2010;4:26–33.
    1. Lassmann H, Schmied M, Vass K, Hickey WF. Bone marrow derived elements and resident microglia in brain inflammation. Glia. 1993;7:19–24.
    1. Ginhoux F, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330:841–845.
    1. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308:1314–1318.
    1. Bechmann I, et al. Immune surveillance of mouse brain perivascular spaces by blood-borne macrophages. Eur J Neurosci. 2001;14:1651–1658.
    1. Mantovani A, Sica A, Locati M. Macrophage polarization comes of age. Immunity. 2005;23:344–346.
    1. Rao KN, Brown MA. Mast cells: multifaceted immune cells with diverse roles in health and disease. Ann N Y Acad Sci. 2008;1143:83–104.
    1. Tanaka R, et al. Migration of enhanced green fluorescent protein expressing bone marrow-derived microglia/macrophage into the mouse brain following permanent focal ischemia. Neuroscience. 2003;117:531–539.
    1. Geissmann F, et al. Development of monocytes, macrophages, and dendritic cells. Science. 2010;327:656–661.
    1. Reichmann G, Schroeter M, Jander S, Fischer HG. Dendritic cells and dendritic-like microglia in focal cortical ischemia of the mouse brain. J Neuroimmunol. 2002;129:125–132.
    1. Borregaard N. Neutrophils, from marrow to microbes. Immunity. 2010;33:657–670.
    1. Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172:2731–2738.
    1. Kreisel D, et al. Mouse vascular endothelium activates CD8+ T lymphocytes in a B7-dependent fashion. J Immunol. 2002;169:6154–6161.
    1. Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–548.
    1. Wan YY. Multi-tasking of helper T cells. Immunology. 2010;130:166–171.
    1. Biron CA, Nguyen KB, Pien GC, Cousens LP, Salazar-Mather TP. Natural killer cells in antiviral defense: function and regulation by innate cytokines. Annu Rev Immunol. 1999;17:189–220.
    1. Bonneville M, O'Brien RL, Born WK. γδ T cell effector functions: a blend of innate programming and acquired plasticity. Nat Rev Immunol. 2010;10:467–478.
    1. Hayday AC. γδ T cells and the lymphoid stress-surveillance response. Immunity. 2009;31:184–196.
    1. Selim MH, Ratan RR. The role of iron neurotoxicity in ischemic stroke. Ageing Res Rev. 2004;3:345–353.
    1. Yanamadala V, Friedlander RM. Complement in neuroprotection and neurodegeneration. Trends Mol Med. 2010;16:69–76.
    1. Rosenbaum DM, et al. Fas (CD95/APO-1) plays a role in the pathophysiology of focal cerebral ischemia. J Neurosci Res. 2000;61:686–692.
    1. Strasser A, Jost PJ, Nagata S. The many roles of FAS receptor signaling in the immune system. Immunity. 2009;30:180–192.
    1. Cui M, et al. Blocking TRAIL-DR5 signaling with soluble DR5 reduces delayed neuronal damage after transient global cerebral ischemia. Neurobiol Dis. 2010;39:138–147.
    1. Martin-Villalba A, et al. CD95 ligand (Fas-L/APO-1L) and tumor necrosis factor-related apoptosis-inducing ligand mediate ischemia-induced apoptosis in neurons. J Neurosci. 1999;19:3809–3817.
    1. Liesz A, et al. Inhibition of lymphocyte trafficking shields the brain against deleterious neuroinflammation after stroke. Brain : a journal of neurology. 2011;134:704–720.
    1. Jauch EC, et al. Association of serial biochemical markers with acute ischemic stroke: the National Institute of Neurological Disorders and Stroke recombinant tissue plasminogen activator Stroke Study. Stroke. 2006;37:2508–2513.
    1. Bornstein NM, et al. Antibodies to brain antigens following stroke. Neurology. 2001;56:529–530.
    1. Dambinova SA, et al. Blood test detecting autoantibodies to N-methyl-D-aspartate neuroreceptors for evaluation of patients with transient ischemic attack and stroke. Clin Chem. 2003;49:1752–1762.
    1. Herrmann M, Vos P, Wunderlich MT, de Bruijn CH, Lamers KJ. Release of glial tissue-specific proteins after acute stroke: A comparative analysis of serum concentrations of protein S-100B and glial fibrillary acidic protein. Stroke. 2000;31:2670–2677.
    1. Missler U, Wiesmann M, Friedrich C, Kaps M. S-100 protein and neuron-specific enolase concentrations in blood as indicators of infarction volume and prognosis in acute ischemic stroke. Stroke. 1997;28:1956–1960.
    1. Rocklin RE, Sheremata WA, Feldman RG, Kies MW, David JR. The Guillain-Barré syndrome and multiple sclerosis. In vitro cellular responses to nervous-tissue antigens. N Engl J Med. 1971;284:803–808.
    1. Gordon PH, et al. Efficacy of minocycline in patients with amyotrophic lateral sclerosis: a phase III randomised trial. Lancet neurology. 2007;6:1045–1053.
    1. Chen Y, et al. Mucosal tolerance to E-selectin provides cell-mediated protection against ischemic brain injury. Proc Natl Acad Sci U S A. 2003;100:15107–15112.
    1. Dirnagl U, Becker K, Meisel A. Preconditioning and tolerance against cerebral ischaemia: from experimental strategies to clinical use. Lancet neurology. 2009;8:398–412.
    1. Koch S, Katsnelson M, Dong C, Perez-Pinzon M. Remote Ischemic Limb Preconditioning After Subarachnoid Hemorrhage: A Phase Ib Study of Safety and Feasibility. Stroke; a journal of cerebral circulation. 2011
    1. Lee SJ, Benveniste EN. Adhesion molecule expression and regulation on cells of the central nervous system. Journal Neuroimmunol. 1999;98:77–88.
    1. Schilling M, Strecker JK, Ringelstein EB, Kiefer R, Schabitz WR. Turn-over of meningeal and perivascular macrophages in the brain of MCP-1-, CCR-2- or double knockout mice. Exp Neurol. 2009;219:583–585.
    1. Terao S, et al. Blood cell-derived RANTES mediates cerebral microvascular dysfunction, inflammation, and tissue injury after focal ischemia-reperfusion. Stroke. 2008;39:2560–2570.
    1. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–832.
    1. Di Virgilio F. Liaisons dangereuses: P2X(7) and the inflammasome. Trends Pharmacol Sci. 2007;28:465–472.
    1. Tesco G, et al. Depletion of GGA3 stabilizes BACE and enhances beta-secretase activity. Neuron. 2007;54:721–737.
    1. Kunz A, et al. Nuclear factor-κB activation and postischemic inflammation are suppressed in CD36-null mice after middle cerebral artery occlusion. J Neurosci. 2008;28:1649–1658.
    1. Arumugam TV, Granger DN, Mattson MP. Stroke and T-cells. Neuromolecular Med. 2005;7:229–242.
    1. Brait VH, et al. Mechanisms contributing to cerebral infarct size after stroke: gender, reperfusion, T lymphocytes, and Nox2-derived superoxide. J Cereb Blood Flow Metab. 2010;30:1306–1317.
    1. Koizumi S, et al. UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis. Nature. 2007;446:1091–1095.
    1. Napoli I, Neumann H. Microglial clearance function in health and disease. Neuroscience. 2009;158:1030–1038.
    1. Hanayama R, et al. Autoimmune disease and impaired uptake of apoptotic cells in MFG-E8-deficient mice. Science. 2004;304:1147–1150.
    1. Strle K, et al. Interleukin-10 in the brain. Crit Rev Immunol. 2001;21:427–449.
    1. Buisson A, et al. Up-regulation of a serine protease inhibitor in astrocytes mediates the neuroprotective activity of transforming growth factor β1. FASEB J. 1998;12:1683–1691.
    1. Grilli M, et al. Interleukin-10 modulates neuronal threshold of vulnerability to ischaemic damage. Eur J Neurosci. 2000;12:2265–2272.
    1. Arai K, Jin G, Navaratna D, Lo EH. Brain angiogenesis in developmental and pathological processes: neurovascular injury and angiogenic recovery after stroke. Febs J. 2009;276:4644–4652.

Source: PubMed

3
구독하다