J. Rha and J. L. Saver, The impact of recanalization on ischemic stroke outcome: a meta-analysis, Stroke, vol.38, pp.967-73, 2007.

U. Dirnagl, C. Iadecola, and M. A. Moskowitz, Pathobiology of ischaemic stroke: an integrated view, Trends Neurosci, vol.22, pp.391-398, 1999.

C. R. Carpenter, S. M. Keim, W. K. Milne, W. J. Meurer, and W. G. Barsan, Best Evidence in Emergency Medicine Investigator Group. Thrombolytic therapy for acute ischemic stroke beyond three hours, J Emerg Med, vol.40, pp.82-92, 2011.

R. G. Nogueira, A. P. Jadhav, D. C. Haussen, A. Bonafe, R. F. Budzik et al., Thrombectomy 6 to 24 Hours after Stroke with a Mismatch between Deficit and Infarct, N Engl J Med, vol.378, pp.11-21, 2018.

A. Drieu, D. Levard, D. Vivien, and M. Rubio, Anti-inflammatory treatments for stroke: from bench to bedside, Ther Adv Neurol Disord, vol.11, p.1756286418789854, 2018.

M. Gauberti, S. Martinez-de-lizarrondo, C. Orset, and D. Vivien, Lack of secondary microthrombosis after thrombin-induced stroke in mice and non-human primates, J Thromb Haemost, vol.12, pp.409-423, 2014.

W. Zhou, A. Liesz, H. Bauer, C. Sommer, B. Lahrmann et al., Postischemic brain infiltration of leukocyte subpopulations differs among murine permanent and transient focal cerebral ischemia models, Brain Pathol, vol.23, pp.34-44, 2013.

M. Gelderblom, F. Leypoldt, K. Steinbach, D. Behrens, C. Choe et al., Temporal and Spatial Dynamics of Cerebral Immune Cell Accumulation in Stroke, Stroke, vol.40, pp.1849-57, 2009.

H. X. Chu, H. A. Kim, S. Lee, J. P. Moore, C. T. Chan et al., Immune cell infiltration in malignant middle cerebral artery infarction: comparison with transient cerebral ischemia, J Cereb Blood Flow Metab, vol.34, pp.450-459, 2014.

C. Orset, R. Macrez, A. R. Young, D. Panthou, E. Angles-cano et al., Mouse model of in situ thromboembolic stroke and reperfusion, Stroke, vol.38, pp.2771-2779, 2007.

L. Behot, A. Gauberti, M. Martinez-de-lizarrondo, S. Montagne, A. Lemarchand et al., GpIb?-VWF blockade restores vessel patency by dissolving platelet aggregates formed under very high shear rate in mice, Blood, vol.123, pp.3354-63, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02153395

K. Kimura, Y. Sakamoto, Y. Iguchi, and K. Shibazaki, Serial changes in ischemic lesion volume and neurological recovery after t-PA therapy, J Neurol Sci, vol.304, pp.35-44, 2011.

J. Pialat, M. Wiart, N. Nighoghossian, P. Adeleine, L. Derex et al., Evolution of lesion volume in acute stroke treated by intravenous t-PA, J Magn Reson Imaging, vol.22, pp.23-31, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00443398

, Effect of intravenous recombinant tissue plasminogen activator on ischemic stroke lesion size measured by computed tomography. NINDS; The National Institute of Neurological Disorders and Stroke (NINDS) rt-PA Stroke Study Group, Stroke, vol.31, pp.2912-2921, 2000.

C. Orset, B. Haelewyn, S. M. Allan, S. Ansar, F. Campos et al., Efficacy of Alteplase in a Mouse Model of Acute Ischemic Stroke: A Retrospective Pooled Analysis, Stroke, vol.47, pp.1312-1320, 2016.

R. Macrez, P. Obiang, M. Gauberti, B. Roussel, A. Baron et al., Antibodies preventing the interaction of tissue-type plasminogen activator with N-methyl-D-aspartate receptors reduce stroke damages and extend the therapeutic window of thrombolysis, Stroke, vol.42, pp.2315-2337, 2011.

S. Suzuki, K. Tanaka, and N. Suzuki, Ambivalent aspects of interleukin-6 in cerebral ischemia: inflammatory versus neurotrophic aspects, J Cereb Blood Flow Metab, vol.29, pp.464-79, 2009.

Y. Yang, H. Liu, H. Zhang, Q. Ye, J. Wang et al., ST2/IL-33-Dependent Microglial Response Limits Acute Ischemic Brain Injury, J Neurosci, vol.37, pp.4692-704, 2017.

X. Liu, J. Liu, S. Zhao, H. Zhang, W. Cai et al., Interleukin-4 Is Essential for Microglia/Macrophage M2 Polarization and Long-Term Recovery After Cerebral Ischemia, Stroke, vol.47, pp.498-504, 2016.

G. Szalay, B. Martinecz, N. Lénárt, Z. Környei, B. Orsolits et al., Microglia protect against brain injury and their selective elimination dysregulates neuronal network activity after stroke, Nature Communications, vol.7, p.11499, 2016.

M. Lalancette-hébert, G. Gowing, A. Simard, Y. C. Weng, and J. Kriz, Selective ablation of proliferating microglial cells exacerbates ischemic injury in the brain, J Neurosci, vol.27, pp.2596-605, 2007.

Y. Lampl, M. Boaz, R. Gilad, M. Lorberboym, R. Dabby et al., Minocycline treatment in acute stroke: an open-label, evaluator-blinded study, Neurology, vol.69, pp.1404-1414, 2007.

P. Srivastava, M. V. Bhasin, A. Bhatia, R. Garg, A. Gaikwad et al., Efficacy of minocycline in acute ischemic stroke: a single-blinded, placebo-controlled trial, Neurol India, vol.60, pp.23-31, 2012.

E. Kohler, D. A. Prentice, T. R. Bates, G. J. Hankey, A. Claxton et al., Intravenous minocycline in acute stroke: a randomized, controlled pilot study and meta-analysis, Stroke, vol.44, pp.2493-2502, 2013.

D. Davalos, J. K. Ryu, M. Merlini, K. M. Baeten, L. Moan et al., Fibrinogeninduced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation, Nat Commun, vol.3, p.1227, 2012.

T. G. References-1.-jovin, A. Chamorro, and E. Cobo, Thrombectomy within 8 hours after symptom onset in ischemic stroke, N Engl J Med, vol.372, pp.2296-2306, 2015.

R. G. Nogueira, A. P. Jadhav, and D. C. Haussen, Thrombectomy 6 to 24 hours after stroke with a mismatch between deficit and infarct, N Engl J Med, vol.378, pp.11-21, 2018.

G. W. Albers, M. P. Marks, and S. Kemp, Thrombectomy for stroke at 6 to 16 hours with selection by perfusion imaging, N Engl J Med, vol.378, pp.708-718, 2018.

S. C. Fagan, L. E. Cronic, and D. C. Hess, Minocycline development for acute ischemic stroke, Transl Stroke Res, vol.2, pp.202-208, 2011.

C. Iadecola and J. Anrather, The immunology of stroke: from mechanisms to translation, Nat Med, vol.17, pp.796-808, 2011.

D. W. Choi, Glutamate neurotoxicity in cortical cell culture is calcium dependent, Neurosci Lett, vol.58, pp.293-297, 1985.

K. Koistinaho and J. , Interactions between Alzheimer's disease and cerebral ischemia-focus on inflammation, Brain Res Brain Res Rev, vol.48, pp.240-250, 2005.

Y. Miyaji, S. Yoshimura, and N. Sakai, Effect of edaravone on favorable outcome in patients with acute cerebral large vessel occlusion: subanalysis of RESCUE-Japan Registry, Neurol Med Chir (Tokyo), vol.55, pp.241-247, 2015.

C. Perego, S. Fumagalli, D. Simoni, and M. , Temporal pattern of expression and colocalization of microglia/macrophage phenotype markers following brain ischemic injury in mice, J Neuroinflammation, vol.8, p.174, 2011.

J. Huang, U. M. Upadhyay, and R. J. Tamargo, Inflammation in stroke and focal cerebral ischemia, Surg Neurol, vol.66, pp.232-245, 2006.

W. A. Muller, Leukocyte-endothelial cell interactions in the inflammatory response, Lab Investig J Tech Methods Pathol, vol.82, pp.521-533, 2002.

B. Engelhardt and R. M. Ransohoff, The ins and outs of T-lymphocyte trafficking to the CNS: anatomical sites and molecular mechanisms, Trends Immunol, vol.26, pp.485-495, 2005.

H. K. Eltzschig and T. Eckle, Ischemia and reperfusion-from mechanism to translation, Nat Med, vol.17, pp.1391-1401, 2011.

M. Gelderblom, F. Leypoldt, and K. Steinbach, Temporal and spatial dynamics of cerebral immune cell accumulation in stroke, Stroke, vol.40, pp.1849-1857, 2009.

H. Mena, D. Cadavid, and E. J. Rushing, Human cerebral infarct: a proposed histopathologic classification based on 137 cases, Acta Neuropathol (Berl), vol.108, pp.524-530, 2004.

W. Zhou, A. Liesz, and H. Bauer, Postischemic brain infiltration of leukocyte subpopulations differs among murine permanent and transient focal cerebral ischemia models: infiltration differs among cerebral ischemia, Brain Pathol, vol.23, pp.34-44, 2013.

M. G. Lansberg, M. Straka, and S. Kemp, MRI profile and response to endovascular reperfusion after stroke (DEFUSE 2): a prospective cohort study, Lancet Neurol, vol.11, pp.860-867, 2012.

J. Elkins, Safety and efficacy of intravenous natalizumab in acute ischemic stroke, 2018.

S. Martinez-de-lizarrondo, C. Gakuba, and B. A. Herbig, Potent thrombolytic effect of n-acetylcysteine on arterial thrombi, Circulation, vol.136, pp.646-660, 2017.

C. Orset, B. Haelewyn, and S. M. Allan, Efficacy of alteplase in a mouse model of acute ischemic stroke: a retrospective pooled analysis, Stroke, vol.47, pp.1312-1318, 2016.

C. Orset, R. Macrez, and A. R. Young, Mouse model of in situ thromboembolic stroke and reperfusion, Stroke, vol.38, pp.2771-2778, 2007.

K. Hossmann, The two pathophysiologies of focal brain ischemia: implications for translational stroke research, J Cereb Blood Flow Metab Off J Int Soc Cereb Blood Flow Metab, vol.32, pp.1310-1316, 2012.

V. E. O'collins, M. R. Macleod, and G. A. Donnan,

, Ann Neurol, vol.59, pp.467-477, 2006.

M. S. Spychala, P. Honarpisheh, and L. D. Mccullough, Sex differences in neuroinflammation and neuroprotection in ischemic stroke, J Neurosci Res, vol.95, issue.1-2, pp.462-471, 2017.

F. Ginhoux, M. Greter, and M. Leboeuf, Fate mapping analysis reveals that adult microglia derive from primitive macrophages, Science, vol.330, pp.841-845, 2010.

G. Yilmaz and D. N. Granger, Leukocyte recruitment and ischemic brain injury, Neuro Molecular Med, vol.12, pp.193-204, 2010.

B. Bmi, Mass Index; AUDIT, Alcohol Use Disorders Identification Test

A. Abtin, R. Jain, A. J. Mitchell, B. Roediger, A. J. Brzoska et al., Perivascular macrophages mediate neutrophil recruitment during bacterial skin infection, Nat Immunol, vol.15, pp.45-53, 2014.

B. O. Fabriek, E. S. Van-haastert, I. Galea, M. M. Polfliet, E. D. Döpp et al., , 2005.

, CD163-positive perivascular macrophages in the human CNS express molecules for antigen recognition and presentation, Glia, vol.51, pp.297-305

G. Faraco, Y. Sugiyama, D. Lane, L. Garcia-bonilla, H. Chang et al., Perivascular macrophages mediate the neurovascular and cognitive dysfunction associated with hypertension, Journal of Clinical Investigation, vol.126, pp.4674-4689, 2016.

A. R. Filipowicz, C. M. Mcgary, G. E. Holder, A. A. Lindgren, E. M. Johnson et al., Proliferation of Perivascular Macrophages Contributes to the Development of Encephalitic Lesions in HIV-Infected Humans and in SIV-Infected Macaques, 2016.

T. Goldmann, P. Wieghofer, M. J. Jordão, F. Prutek, N. Hagemeyer et al., Origin, fate and dynamics of macrophages at central nervous system interfaces, Nature Immunology, vol.17, pp.797-805, 2016.

A. K. Harris, A. Ergul, A. Kozak, L. S. Machado, M. H. Johnson et al., Effect of neutrophil depletion on gelatinase expression, edema formation and hemorrhagic transformation after focal ischemic stroke, BMC Neurosci, vol.6, p.49, 2005.

H. He, J. J. Mack, E. Güç, C. M. Warren, M. L. Squadrito et al., Perivascular Macrophages Limit Permeability, Arterioscler. Thromb. Vasc. Biol, vol.36, pp.2203-2212, 2016.

S. Kida, P. V. Steart, E. T. Zhang, and R. O. Weller, Perivascular cells act as scavengers in the cerebral perivascular spaces and remain distinct from pericytes, microglia and macrophages, Acta Neuropathol, vol.85, pp.646-652, 1993.

E. Lemarchand, M. Gauberti, S. Martinez-de-lizarrondo, H. Villain, Y. Repessé et al., Impact of alcohol consumption on the outcome of ischemic stroke and thrombolysis: role of the hepatic clearance of tissue-type plasminogen activator, Stroke, vol.46, pp.1641-1650, 2015.

L. Mendes-jorge, D. Ramos, M. Luppo, C. Llombart, G. Alexandre-pires et al., Scavenger function of resident autofluorescent perivascular macrophages and their contribution to the maintenance of the blood-retinal barrier, Invest. Ophthalmol. Vis. Sci, vol.50, pp.5997-6005, 2009.

K. Möller, C. Pösel, A. Kranz, I. Schulz, J. Scheibe et al., Arterial Hypertension Aggravates Innate Immune Responses after Experimental Stroke, Front Cell Neurosci, vol.9, p.461, 2015.

J. Neumann, M. Riek-burchardt, J. Herz, T. R. Doeppner, R. König et al., Very-late-antigen-4 (VLA-4)-mediated brain invasion by neutrophils leads to interactions with microglia, increased ischemic injury and impaired behavior in experimental stroke, Acta Neuropathol, vol.129, pp.259-277, 2015.

M. J. O'donnell, D. Xavier, L. Liu, H. Zhang, S. L. Chin et al., Risk factors for ischaemic and intracerebral haemorrhagic stroke in 22 countries (the INTERSTROKE study): a case-control study, Lancet, vol.376, pp.112-123, 2010.

G. C. Olivera, X. Ren, S. K. Vodnala, J. Lu, L. Coppo et al., Nitric Oxide Protects against Infection-Induced Neuroinflammation by Preserving the Stability of the Blood-Brain Barrier, PLOS Pathogens, vol.12, p.1005442, 2016.

C. Orset, R. Macrez, A. R. Young, D. Panthou, E. Angles-cano et al., Mouse model of in situ thromboembolic stroke and reperfusion, Stroke, vol.38, pp.2771-2778, 2007.

C. Orset, B. Haelewyn, S. M. Allan, S. Ansar, F. Campos et al., Efficacy of Alteplase in a Mouse Model of Acute Ischemic Stroke: A Retrospective Pooled Analysis, Stroke, vol.47, pp.1312-1318, 2016.

L. Park, K. Uekawa, L. Garcia-bonilla, K. Koizumi, M. Murphy et al., Brain Perivascular Macrophages Initiate the Neurovascular Dysfunction of Alzheimer A? Peptides, Circ. Res, vol.121, pp.258-269, 2017.

J. Pedragosa, A. Salas-perdomo, M. Gallizioli, R. Cugota, F. Miró-mur et al., CNS-border associated macrophages respond to acute ischemic stroke attracting granulocytes and promoting vascular leakage, Acta Neuropathol Commun, vol.6, p.76, 2018.

A. Quenault, S. Martinez-de-lizarrondo, O. Etard, M. Gauberti, C. Orset et al., Molecular magnetic resonance imaging discloses endothelial activation after transient ischaemic attack, Brain, vol.140, pp.146-157, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01431517

A. Schmidt, J. Strecker, S. Hucke, N. Bruckmann, M. Herold et al., Targeting Different Monocyte/Macrophage Subsets Has No Impact on Outcome in Experimental Stroke, Stroke, vol.48, pp.1061-1069, 2017.

, CLO-treated mice 24 hours after stroke onset. (D) Representative photomicrographs of VCAM1

B. M. Bettcher and J. H. Kramer, Longitudinal inflammation, cognitive decline, and Alzheimer's disease: a minireview, Clin. Pharmacol. Ther, vol.96, pp.464-469, 2014.

D. L. Brody, M. Donald, C. Kessens, C. C. Yuede, C. Parsadanian et al., Electromagnetic controlled cortical impact device for precise, graded experimental traumatic brain injury, J. Neurotrauma, vol.24, pp.657-673, 2007.

T. Cao, T. C. Thomas, J. M. Ziebell, J. R. Pauly, and J. Lifshitz, Morphological and genetic activation of microglia after diffuse traumatic brain injury in the rat, Neuroscience, vol.225, pp.65-75, 2012.

J. D. Cassidy, L. J. Carroll, P. M. Peloso, J. Borg, V. Holst et al., Incidence, risk factors and prevention of mild traumatic brain injury: results of the WHO Collaborating Centre Task Force on Mild Traumatic Brain Injury, J Rehabil Med, pp.28-60, 2004.

J. M. Coughlin, Y. Wang, C. A. Munro, S. Ma, C. Yue et al., Neuroinflammation and brain atrophy in former NFL players: An in vivo multimodal imaging pilot study, Neurobiol. Dis, vol.74, pp.58-65, 2015.

M. Cruz-haces, J. Tang, G. Acosta, J. Fernandez, and R. Shi, Pathological correlations between traumatic brain injury and chronic neurodegenerative diseases, Transl Neurodegener, vol.6, p.20, 2017.

M. C. Dewan, A. Rattani, S. Gupta, R. E. Baticulon, Y. Hung et al., Estimating the global incidence of traumatic brain injury, J. Neurosurg, vol.2018, pp.1-18

C. K. Donat, K. Gaber, J. Meixensberger, P. Brust, L. H. Pinborg et al., Changes in Binding

. Clinde and . High, Affinity Translocator Protein 18 kDa (TSPO) Selective Radioligand in a Rat Model of Traumatic Brain Injury, Neuromolecular Med, vol.18, pp.158-169, 2016.

C. K. Donat, G. Scott, S. M. Gentleman, and M. Sastre, Microglial Activation in Traumatic Brain Injury, Front Aging Neurosci, vol.9, p.208, 2017.

B. Fehily and M. Fitzgerald, Repeated Mild Traumatic Brain Injury: Potential Mechanisms of Damage, Cell Transplant, vol.26, pp.1131-1155, 2017.

H. Folkersma, R. Boellaard, M. Yaqub, R. W. Kloet, A. D. Windhorst et al., Widespread and prolonged increase in (R)-(11)C-PK11195 binding after traumatic brain injury, J. Nucl. Med, vol.52, pp.1235-1239, 2011.

L. Foucault-fruchard, A. Doméné, G. Page, M. Windsor, P. Emond et al., Neuroprotective effect of the alpha 7 nicotinic receptor agonist PHA 543613 in an in vivo excitotoxic adult rat model, Neuroscience, vol.356, pp.52-63, 2017.

A. P. Fournier, A. Quenault, S. Martinez-de-lizarrondo, M. Gauberti, G. Defer et al., Prediction of disease activity in models of multiple sclerosis by molecular magnetic resonance imaging of P-selectin, Proc. Natl. Acad. Sci. U.S.A, vol.114, pp.6116-6121, 2017.

R. C. Gardner and K. Yaffe, Epidemiology of mild traumatic brain injury and neurodegenerative disease, Mol. Cell. Neurosci, vol.66, pp.75-80, 2015.

M. Gauberti, A. P. Fournier, F. Docagne, D. Vivien, and S. Martinez-de-lizarrondo, Molecular Magnetic Resonance Imaging of Endothelial Activation in the Central Nervous System, Theranostics, vol.8, pp.1195-1212, 2018.

M. Gauberti, A. Montagne, O. A. Marcos-contreras, L. Béhot, A. Maubert et al., Ultra-sensitive molecular MRI of vascular cell adhesion molecule-1 reveals a dynamic inflammatory penumbra after strokes, Stroke, vol.44, pp.1988-1996, 2013.

R. Grossman, C. M. Paden, P. A. Fry, R. S. Rhodes, and A. Biegon, Persistent region-dependent neuroinflammation, NMDA receptor loss and atrophy in an animal model of penetrating brain injury, Future Neurol, vol.7, pp.329-339, 2012.

A. Henn, S. Kirner, and M. Leist, TLR2 hypersensitivity of astrocytes as functional consequence of previous inflammatory episodes, J. Immunol, vol.186, pp.3237-3247, 2011.

E. Hennessy, É. W. Griffin, and C. Cunningham, Astrocytes Are Primed by Chronic Neurodegeneration to Produce Exaggerated Chemokine and Cell Infiltration Responses to Acute Stimulation with the Cytokines IL-1? and TNF-?, J. Neurosci, vol.35, pp.8411-8422, 2015.

A. P. Hermida, W. M. Mcdonald, K. Steenland, and A. Levey, The association between late-life depression, mild cognitive impairment and dementia: is inflammation the missing link?, Expert Rev Neurother, vol.12, pp.1339-1350, 2012.

M. M. Hughes, R. H. Field, V. H. Perry, C. L. Murray, and C. Cunningham, Microglia in the degenerating brain are capable of phagocytosis of beads and of apoptotic cells, but do not efficiently remove PrPSc, even upon LPS stimulation, Glia, vol.58, pp.2017-2030, 2010.

I. Israel, A. Ohsiek, E. Al-momani, A. , C. Stetter et al.,

F. Dpa, -714 micro-positron emission tomography and autoradiography imaging of microglia activation after closed head injury in mice, J Neuroinflammation, vol.13, p.140, 2016.

Z. Kikinis, M. Muehlmann, O. Pasternak, S. Peled, P. Kulkarni et al., Diffusion imaging of mild traumatic brain injury in the impact accelerated rodent model: A pilot study, Brain Inj, vol.31, pp.1376-1381, 2017.

T. Madsen, A. Erlangsen, S. Orlovska, R. Mofaddy, M. Nordentoft et al., Association Between Traumatic Brain Injury and Risk of Suicide, JAMA, vol.320, pp.580-588, 2018.

M. A. Mcateer, A. M. Akhtar, C. Von-zur-muhlen, and R. P. Choudhury, An approach to molecular imaging of atherosclerosis, thrombosis, and vascular inflammation using microparticles of iron oxide, Atherosclerosis, vol.209, pp.18-27, 2010.

A. A. Miller and S. J. Spencer, Obesity and neuroinflammation: a pathway to cognitive impairment, Brain Behav. Immun, vol.42, pp.10-21, 2014.

A. Montagne, M. Gauberti, R. Macrez, A. Jullienne, A. Briens et al., Ultra-sensitive molecular MRI of cerebrovascular cell activation enables early detection of chronic central nervous system disorders, NeuroImage, vol.63, pp.760-770, 2012.

D. M. Norden, M. M. Muccigrosso, and J. P. Godbout, Microglial priming and enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and neurodegenerative disease, Neuropharmacology, vol.96, pp.29-41, 2015.

P. Obiang, R. Macrez, A. Jullienne, T. Bertrand, F. Lesept et al., GluN2D subunit-containing NMDA receptors control tissue plasminogen activator-mediated spatial memory, J. Neurosci, vol.32, pp.12726-12734, 2012.

D. C. Perry, V. E. Sturm, M. J. Peterson, C. F. Pieper, T. Bullock et al., Association of traumatic brain injury with subsequent neurological and psychiatric disease: a meta-analysis, J. Neurosurg, vol.124, pp.511-526, 2016.

V. H. Perry and C. Holmes, Microglial priming in neurodegenerative disease, Nat Rev Neurol, vol.10, pp.217-224, 2014.

M. Pruvost, M. Lépine, C. Leonetti, O. Etard, M. Naveau et al., ADAMTS-4 in oligodendrocytes contributes to myelination with an impact on motor function, Glia, vol.65, pp.1961-1975, 2017.

A. Quenault, S. Martinez-de-lizarrondo, O. Etard, M. Gauberti, C. Orset et al., Molecular magnetic resonance imaging discloses endothelial activation after transient ischaemic attack, Brain, vol.140, pp.146-157, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01431517

V. Ramaglia, T. R. Hughes, R. M. Donev, M. M. Ruseva, X. Wu et al., C3-dependent mechanism of microglial priming relevant to multiple sclerosis, Proc. Natl. Acad. Sci. U.S.A, vol.109, pp.965-970, 2012.

A. F. Ramlackhansingh, D. J. Brooks, R. J. Greenwood, S. K. Bose, F. E. Turkheimer et al., Inflammation after trauma: microglial activation and traumatic brain injury, Ann. Neurol, vol.70, pp.374-383, 2011.

M. T. Schwalm, M. Pasquali, S. P. Miguel, D. Santos, J. Vuolo et al., Acute brain inflammation and oxidative damage are related to long-term cognitive deficits and markers of neurodegeneration in sepsis-survivor rats, Mol. Neurobiol, vol.49, pp.380-385, 2014.

L. Siebold, A. Obenaus, and R. Goyal, Criteria to define mild, moderate, and severe traumatic brain injury in the mouse controlled cortical impact model, Exp. Neurol, vol.310, pp.48-57, 2018.

N. Sollmann, P. S. Echlin, V. Schultz, P. V. Viher, A. E. Lyall et al., Sex differences in white matter alterations following repetitive subconcussive head impacts in collegiate ice hockey players, Neuroimage Clin, vol.17, pp.642-649, 2018.

G. Teasdale and B. Jennett, Assessment of coma and impaired consciousness. A practical scale, Lancet, 1974.

S. Alfonso-loeches, M. Pascual-lucas, A. M. Blanco, I. Sanchez-vera, and C. Guerri, Pivotal Role of TLR4 Receptors in Alcohol-Induced Neuroinflammation and Brain Damage, Journal of Neuroscience, vol.30, pp.8285-8295, 2010.

I. C. Anthony, D. H. Crawford, and J. E. Bell, B lymphocytes in the normal brain: contrasts with HIV-associated lymphoid infiltrates and lymphomas, Brain, vol.126, pp.1058-1067, 2003.

A. Arac, M. A. Grimbaldeston, A. R. Nepomuceno, O. Olayiwola, M. P. Pereira et al., Evidence that Meningeal Mast Cells Can Worsen Stroke Pathology in Mice, The American Journal of Pathology, vol.184, pp.2493-2504, 2014.

R. C. Armstrong, A. J. Mierzwa, C. M. Marion, and G. M. Sullivan, White matter involvement after TBI: Clues to axon and myelin repair capacity, Exp. Neurol. 275 Pt, vol.3, pp.328-333, 2016.

M. J. Attella, A. Nattinville, and D. G. Stein, Hormonal state affects recovery from frontal cortex lesions in adult female rats, Behav. Neural Biol, vol.48, pp.352-367, 1987.

J. Audoy-rémus, J. Richard, D. Soulet, H. Zhou, P. Kubes et al., Rod-Shaped monocytes patrol the brain vasculature and give rise to perivascular macrophages under the influence of proinflammatory cytokines and angiopoietin-2, J. Neurosci, vol.28, pp.10187-10199, 2008.

A. D. Bachstetter, B. Xing, L. De-almeida, E. R. Dimayuga, D. M. Watterson et al., oi fla ato toki e up-regulation induced by toll-like receptor (TLR) ligands or beta-a loid A?, p.79, 2011.

A. D. Ba-hstette, R. K. Ro-e, M. Ka-eko, D. Gouldi-g, J. Lifshitz et al., The p ? MAPK Regulates Microglial Responsiveness to Diffuse Traumatic Brain Injury, J. Neurosci, vol.33, pp.6143-6153

V. Banwell, E. S. Sena, and M. R. Macleod, Systematic review and stratified meta-analysis of the efficacy of interleukin-1 receptor antagonist in animal models of stroke, J Stroke Cerebrovasc Dis, vol.18, pp.269-276, 2009.

I. Bechmann, E. Kwidzinski, A. D. Kovac, E. Simbürger, T. Horvath et al., Turnover of Rat Brain Perivascular Cells, Experimental Neurology, vol.168, pp.242-249, 2001.

I. Bechmann, J. Priller, A. Kovac, M. Böntert, T. Wehner et al., Immune surveillance of mouse brain perivascular spaces by blood-borne macrophages, Eur. J. Neurosci, vol.14, pp.1651-1658, 2001.

K. Becker, D. Kindrick, J. Relton, J. Harlan, W. et al., Antibody to the alpha4 integrin decreases infarct size in transient focal cerebral ischemia in rats, Stroke, vol.32, pp.206-211, 2001.

J. I. Beier and C. J. Mcclain, Mechanisms and cell signaling in alcoholic liver disease, Biological Chemistry, vol.391, 2010.

C. Benakis, L. Garcia-bonilla, C. Iadecola, and J. Anrather, The role of microglia and myeloid immune cells in acute cerebral ischemia, Frontiers in Cellular Neuroscience, vol.8, 2015.

M. Bi, A. Gladbach, J. Van-eersel, A. Ittner, M. Przybyla et al., , 2017.

K. Blinzinger and G. Kreutzberg, Displacement of synaptic terminals from regenerating motoneurons by microglial cells, Zeitschrift Für Zellforschung Und Mikroskopische Anatomie, vol.85, pp.145-157, 1968.

P. C. Blumbergs, G. Scott, J. M. Vis, H. Wainwright, D. A. Simpson et al., Topography of Axonal Injury as Defined by Amyloid Precursor Protein and the Sector Scoring Method in Mild and Severe Closed Head Injury, Journal of Neurotrauma, vol.12, pp.565-572, 1995.

C. H. Bombardier, J. R. Fann, N. R. Temkin, P. C. Esselman, J. Barber et al., Rates of major depressive disorder and clinical outcomes following traumatic brain injury, JAMA, vol.303, pp.1938-1945, 2010.

T. Bonnard and C. E. Hagemeyer, Ferric Chloride-induced Thrombosis Mouse Model on Carotid Artery and Mesentery Vessel, J Vis Exp, 2015.

M. P. Bowes, R. Rothlein, S. C. Fagan, and J. A. Zivin, Monoclonal antibodies preventing leukocyte activation reduce experimental neurologic injury and enhance efficacy of thrombolytic therapy, Neurology, vol.45, pp.815-819, 1995.

C. L. Bowman, J. Ding, F. Sachs, and M. Sokabe, Mechanotransducing ion channels in astrocytes, Brain Research, vol.584, pp.272-286, 1992.

S. Bracard, X. Ducrocq, J. L. Mas, M. Soudant, C. Oppenheim et al., Mechanical thrombectomy after intravenous alteplase versus alteplase alone after stroke (THRACE): a randomised controlled trial, Lancet Neurol, vol.15, pp.1138-1147, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01825521

G. C. Brown and J. J. Neher, Microglial phagocytosis of live neurons, Nat. Rev. Neurosci, vol.15, pp.209-216, 2014.

K. D. Browne, X. Chen, D. F. Meaney, and D. H. Smith, Mild traumatic brain injury and diffuse axonal injury in swine, J. Neurotrauma, vol.28, pp.1747-1755, 2011.

J. E. Burda, A. M. Bernstein, and M. V. Sofroniew, Astrocyte roles in traumatic brain injury, Exp. Neurol. 275 Pt, vol.3, pp.305-315, 2016.

T. G. Bush, N. Puvanachandra, C. H. Horner, A. Polito, T. Ostenfeld et al., Leukocyte Infiltration, Neuronal Degeneration, and Neurite Outgrowth after Ablation of Scar-Forming, Reactive Astrocytes in Adult Transgenic Mice. Neuron, vol.23, pp.297-308, 1999.
DOI : 10.1016/s0896-6273(00)80781-3

URL : https://doi.org/10.1016/s0896-6273(00)80781-3

C. Caravagna, A. Jaouën, S. Desplat-jégo, K. K. Fenrich, E. Bergot et al., , 2018.

A. E. Cardona, E. P. Pioro, M. E. Sasse, V. Kostenko, S. M. Cardona et al., Control of microglial neurotoxicity by the fractalkine receptor, Nature Neuroscience, vol.9, pp.917-924, 2006.

T. M. Carlos, R. S. Clark, D. Franicola-higgins, J. K. Schiding, and P. M. Kochanek, Expression of endothelial adhesion molecules and recruitment of neutrophils after traumatic brain injury in rats, J. Leukoc. Biol, vol.61, pp.279-285, 1997.

G. Chen, S. Zhang, J. Shi, J. Ai, M. Qi et al., Simvastatin reduces secondary brain injury caused by cortical contusion in rats: possible involvement of TLR4/NF-kappaB pathway, Exp. Neurol, vol.216, pp.398-406, 2009.

S. Chen, T. Hung, C. Chen, K. Lin, Y. Huang et al., Lovastatin improves histological and functional outcomes and reduces inflammation after experimental traumatic brain injury, Life Sci, vol.81, pp.288-298, 2007.

H. R. Chinnery, M. J. Ruitenberg, and P. G. Mcmenamin, Novel characterization of monocytederived cell populations in the meninges and choroid plexus and their rates of replenishment in bone marrow chimeric mice, J. Neuropathol. Exp. Neurol, vol.69, pp.896-909, 2010.

H. X. Chu, H. A. Kim, S. Lee, J. P. Moore, C. T. Chan et al., Immune cell infiltration in malignant middle cerebral artery infarction: comparison with transient cerebral ischemia, J. Cereb. Blood Flow Metab, vol.34, pp.450-459, 2014.

M. I. Cobb, .. H. Bruce, D. J. Graffagnino, C. Hauck, and E. F. , Symbiotic relationship of IV-tPA and mechanical thrombectomy in a case of acute tandem ICA-MCA occlusion, J Clin Neurosci, vol.38, pp.68-71, 2017.

J. M. Coughlin, Y. Wang, C. A. Munro, S. Ma, C. Yue et al., Neuroinflammation and brain atrophy in former NFL players: An in vivo multimodal imaging pilot study, Neurobiol. Dis, vol.74, pp.58-65, 2015.

A. S. Cunningham, R. Salvador, J. P. Coles, D. A. Chatfield, P. G. Bradley et al., Physiological thresholds for irreversible tissue damage in contusional regions following traumatic brain injury, Brain, vol.128, pp.1931-1942, 2005.

C. Cunningham, D. Boche, and V. H. Perry, Transforming growth factor beta1, the dominant cytokine in murine prion disease: influence on inflammatory cytokine synthesis and alteration of vascular extracellular matrix, Neuropathol. Appl. Neurobiol, vol.28, pp.107-119, 2002.

P. M. D'agosti-o, A. Gottf-ied-blackmore, N. Anandasabapathy, and K. Bulloch, Brain dendritic cells: biology and pathology, Acta Neuropathol, vol.124, pp.599-614, 2012.

S. Daly, M. Thorpe, S. Rockswold, M. Hubbard, T. Bergman et al., , 2018.

, Hyperbaric Oxygen Therapy in the Treatment of Acute Severe Traumatic Brain Injury: A Systematic Review, J. Neurotrauma, vol.35, pp.623-629

A. Deczkowska, K. Baruch, and M. Schwartz, Type I/II Interferon Balance in the Regulation of Brain Physiology and Pathology, Trends in Immunology, vol.37, pp.181-192, 2016.

L. H. Deddens, G. A. Van-tilborg, K. Van-der-marel, H. Hunt, A. Van-der-toorn et al., Vivo Molecular MRI of ICAM-1 Expression on Endothelium and Leukocytes from Subacute to Chronic Stages After Experimental Stroke, 2017.

A. Denes, R. Vidyasagar, J. Feng, J. Narvainen, B. W. Mccoll et al., Proliferating resident microglia after focal cerebral ischaemia in mice, J. Cereb. Blood Flow Metab, vol.27, pp.1941-1953, 2007.

A. Deoke, S. Deoke, A. Saoji, and S. Hajare, Profile of Modifiable and Non-Modifiable Risk Factors in Stroke in a Rural Based Tertiary Care Hospital -A Case Control Study, Glob J Health Sci, vol.4, pp.158-163, 2012.

D. S. Dewitt and D. S. Prough, Blast-induced brain injury and posttraumatic hypotension and hypoxemia, J. Neurotrauma, vol.26, pp.877-887, 2009.

U. Dirnagl, C. Iadecola, and M. A. Moskowitz, Pathobiology of ischaemic stroke: an integrated view, Trends Neurosci, vol.22, pp.391-397, 1999.

C. D'mello and M. G. , I u e-to-Brain Communication Pathways in InflammationAssociated Sickness and Depression, Curr Top Behav Neurosci, vol.31, pp.73-94

C. D'mello, T. Le, and M. G. Swain, Cerebral Microglia Recruit Monocytes into the Brain in Response to Tumor Necrosis Factor Signaling during Peripheral Organ Inflammation, Journal of Neuroscience, vol.29, pp.2089-2102, 2009.

C. D'mello, K. Riazi, T. Le, K. M. Ste-e-s, A. Wa-g et al., , 2013.

, P-selectin-mediated monocyte-cerebral endothelium adhesive interactions link peripheral organ inflammation to sickness behaviors, J. Neurosci, vol.33, pp.14878-14888

M. A. Dobrovolskaia and S. N. Vogel, Toll receptors, CD14, and macrophage activation and deactivation by LPS, Microbes and Infection, vol.4, pp.903-914, 2002.

K. P. Doyle, L. N. Quach, M. Sole, R. C. Axtell, T. V. Nguyen et al., B-Lymphocyte-Mediated Delayed Cognitive Impairment following Stroke, Journal of Neuroscience, vol.35, pp.2133-2145, 2015.

J. P. Dreier, C. Reiffurth, J. Woitzik, J. A. Hartings, C. Drenckhahn et al., How spreading depolarization can be the pathophysiological correlate of both migraine aura and stroke, Acta Neurochir. Suppl, vol.120, pp.137-140, 2015.

A. Drieu, S. Martinez-de-lizarrondo, and M. Rubio, Stopping Inflammation in Stroke: Role of ST2/IL-33 Signaling, J. Neurosci, vol.37, pp.9614-9616, 2017.

A. Drieu, D. Levard, D. Vivien, and M. Rubio, Anti-inflammatory treatments for stroke: from bench to bedside, Ther Adv Neurol Disord, vol.11, p.1756286418789854, 2018.

J. Drouin-ouellet, S. J. Sawiak, G. Cisbani, M. Lagacé, W. Kuan et al., Cerebrovascular and blood-brain barrier impairments in Hu ti gto 's disease: Pote tial i pli atio s fo its pathoph siolog, A . Neu ol, vol.78, pp.160-177, 2015.

A. Ducroquet, D. Leys, A. Saabi, F. Richard, C. Cordonnier et al., Influence of chronic ethanol consumption on the neurological severity in patients with acute cerebral ischemia, Stroke, vol.44, pp.2324-2326, 2013.

W. E. Durrant, D. , and X. , Systemic acquired resistance, Annu Rev Phytopathol, vol.42, pp.185-209, 2004.

B. Engelhardt and R. M. Ransohoff, The ins and outs of T-lymphocyte trafficking to the CNS: anatomical sites and molecular mechanisms, Trends Immunol, vol.26, pp.485-495, 2005.

, Use of anti-ICAM-1 therapy in ischemic stroke: results of the Enlimomab Acute Stroke Trial, Enlimomab Acute Stroke Trial Investigators, vol.57, pp.1428-1434, 2001.

G. Enzmann, C. Mysiorek, R. Gorina, Y. Cheng, S. Ghavampour et al., The neurovascular unit as a selective barrier to polymorphonuclear granulocyte (PMN) infiltration into the brain after ischemic injury, Acta Neuropathol, vol.125, pp.395-412, 2013.

G. U. Enzmann, S. Pavlidou, M. Vaas, J. Klohs, and B. Engelhardt, ICAM-1null C57BL/6 Mice Are Not Protected from Experimental Ischemic Stroke, Transl Stroke Res, 2018.

N. A. Essani, G. M. Mcguire, A. M. Manning, J. , and H. , Differential induction of mRNA for ICAM-1 and selectins in hepatocytes, Kupffer cells and endothelial cells during endotoxemia, Biochem. Biophys. Res. Commun, vol.211, pp.74-82, 1995.

G. Faraco, Y. Sugiyama, D. Lane, L. Garcia-bonilla, H. Chang et al., Perivascular macrophages mediate the neurovascular and cognitive dysfunction associated with hypertension, Journal of Clinical Investigation, vol.126, pp.4674-4689, 2016.

G. Faraco, L. Park, J. Anrather, and C. Iadecola, Brain perivascular macrophages: characterization and functional roles in health and disease, Journal of Molecular Medicine, vol.95, pp.1143-1152, 2017.

A. Farkas and L. Kemény, Alcohol, liver, systemic inflammation and skin: a focus on patients with psoriasis, Skin Pharmacol Physiol, vol.26, pp.119-126, 2013.

D. M. Feeney, M. G. Boyeson, R. T. Linn, H. M. Murray, and W. G. Dail, Responses to cortical injury: I. Methodology and local effects of contusions in the rat, Brain Res, vol.211, pp.67-77, 1981.

V. Felipo, Hepatic encephalopathy: effects of liver failure on brain function, Nature Reviews Neuroscience, vol.14, pp.851-858, 2013.

Y. Feng, S. Liao, C. Wei, D. Jia, K. Wood et al., Infiltration and persistence of lymphocytes during late-stage cerebral ischemia in middle cerebral artery occlusion and photothrombotic stroke models, J Neuroinflammation, vol.14, p.248, 2017.

A. M. Fenn, J. C. Gensel, Y. Huang, P. G. Popovich, J. Lifshitz et al., Immune activation promotes depression 1 month after diffuse brain injury: a role for primed microglia, Biol. Psychiatry, vol.76, pp.575-584, 2014.

A. R. Filipowicz, C. M. Mcgary, G. E. Holder, A. A. Lindgren, E. M. Johnson et al., Proliferation of Perivascular Macrophages Contributes to the Development of Encephalitic Lesions in HIV-Infected Humans and in SIV-Infected Macaques, 2016.

K. Fleischhauer, , 1964.

, Z Zellforsch Mikrosk Anat, vol.64, pp.140-152

C. L. Floyd, F. A. Gorin, and B. G. Lyeth, Mechanical strain injury increases intracellular sodium and reverses Na+/Ca2+ exchange in cortical astrocytes, Glia, vol.51, pp.35-46

J. Flygt, A. Djupsjö, F. Lenne, and N. Marklund, Myelin loss and oligodendrocyte pathology in white matter tracts following traumatic brain injury in the rat, European Journal of Neuroscience, vol.38, pp.2153-2165

E. A. Fontes-júnior, C. S. Maia, L. M. Fernandes, W. Gomes-leal, A. Costa-malaquias et al., Chronic Alcohol Intoxication and Cortical Ischemia: Study of Their Comorbidity and the Protective Effects of Minocycline, Oxid Med Cell Longev, 2016.

I. Galea, K. Palin, T. A. Newman, N. Van-rooijen, V. H. Perry et al., Mannose receptor expression specifically reveals perivascular macrophages in normal, injured, and diseased mouse brain, Glia, vol.49, pp.375-384, 2005.

J. H. Garcia, K. F. Liu, and M. P. Bree, Effects of CD11b/18 monoclonal antibody on rats with permanent middle cerebral artery occlusion, Am. J. Pathol, vol.148, pp.241-248, 1996.

L. Garcia-bonilla, G. Faraco, J. Moore, M. Murphy, G. Racchumi et al., Spatio-temporal profile, phenotypic diversity, and fate of recruited monocytes into the post-ischemic brain, Journal of Neuroinflammation, vol.13, 2016.

L. Garcia-bonilla, D. Brea, C. Benakis, D. Lane, M. Murphy et al., Endogenous protection from ischemic brain injury by preconditioned monocytes, J. Neurosci, 2018.

C. M. Gardiner and K. H. Mills, The cells that mediate innate immune memory and their functional significance in inflammatory and infectious diseases, Semin. Immunol, vol.28, pp.343-350, 2016.

M. Gauberti, A. Montagne, O. A. Marcos-contreras, L. Béhot, A. Maubert et al., , 2013.

, Ultra-sensitive molecular MRI of vascular cell adhesion molecule-1 reveals a dynamic inflammatory penumbra after strokes, Stroke, vol.44, pp.1988-1996

M. Gauberti, S. Martinez-de-lizarrondo, C. Orset, and D. Vivien, Lack of secondary microthrombosis after thrombin-induced stroke in mice and non-human primates, J. Thromb. Haemost, vol.12, pp.409-414, 2014.

M. Gauberti, A. P. Fournier, F. Docagne, D. Vivien, and S. Martinez-de-lizarrondo, Molecular Magnetic Resonance Imaging of Endothelial Activation in the Central Nervous System, Theranostics, vol.8, pp.1195-1212, 2018.

S. Gautier, T. Ouk, O. Petrault, J. Caron, and R. Bordet, Neutrophils contribute to intracerebral haemorrhages after treatment with recombinant tissue plasminogen activator following cerebral ischaemia, Br. J. Pharmacol, vol.156, pp.673-679, 2009.

M. Gelderblom, F. Leypoldt, K. Steinbach, D. Behrens, C. Choe et al., Temporal and Spatial Dynamics of Cerebral Immune Cell Accumulation in Stroke, Stroke, vol.40, pp.1849-1857, 2009.

S. Girard, D. Brough, G. Lopez-castejon, J. Giles, N. J. Rothwell et al., Microglia and macrophages differentially modulate cell death after brain injury caused by oxygen-glucose deprivation in organotypic brain slices: Microglial Phenotype and Brain Injury, Glia, vol.61, pp.813-824, 2013.

T. Goldmann, P. Wieghofer, M. J. Jordão, F. Prutek, N. Hagemeyer et al., Origin, fate and dynamics of macrophages at central nervous system interfaces, Nature Immunology, vol.17, pp.797-805, 2016.

L. B. Goldstein, IV tPA for acute ischemic stroke, 2016.

D. Gomez-nicola and V. H. Perry, Microglial dynamics and role in the healthy and diseased brain: a paradigm of functional plasticity, Neuroscientist, vol.21, pp.169-184, 2015.

R. Go-i-a, M. Fo-t-nie-es, L. Kisi-ousk, and T. Sa-talu-ia, Ast o te TLR activation induces a proinflammatory environment through the interplay between MyD88-dependent NF?B sig ali g, Glia, vol.59, pp.242-255

A. Gottfried-blackmore, U. W. Kaunzner, J. Idoyaga, J. C. Felger, B. S. Mcewen et al., PNAS, vol.106, pp.20918-20923, 2009.

M. B. Graeber, W. J. Streit, and G. W. Kreutzberg, Identity of ED2-positive perivascular cells in rat brain, J. Neurosci. Res, vol.22, pp.103-106, 1989.

M. Greter, I. Lelios, and A. L. Croxford, Microglia Versus Myeloid Cell Nomenclature during Brain Inflammation, Front. Immunol, vol.6, 2015.

J. W. Griffith, C. L. Sokol, and A. D. Luster, Chemokines and Chemokine Receptors: Positioning Cells for Host Defense and Immunity, Annual Review of Immunology, vol.32, pp.659-702, 2014.

V. C. Hachinski, N. A. Lassen, M. , and J. , Multi-infarct dementia. A cause of mental deterioration in the elderly, Lancet, vol.2, pp.207-210, 1974.

N. P. Hailer, I. Bechmann, S. Heizmann, and R. Nitsch, Adhesion molecule expression on phagocytic microglial cells following anterograde degeneration of perforant path axons, Hippocampus, vol.7, pp.341-349, 1997.

E. D. Hall, P. G. Sullivan, T. R. Gibson, K. M. Pavel, B. M. Thompson et al., Spatial and temporal characteristics of neurodegeneration after controlled cortical impact in mice: more than a focal brain injury, J. Neurotrauma, vol.22, pp.252-265, 2005.

U. Hanisch and H. Kettenmann, Microglia: active sensor and versatile effector cells in the normal and pathologic brain, Nature Neuroscience, vol.10, pp.1387-1394, 2007.

A. K. Harris, A. Ergul, A. Kozak, L. S. Machado, M. H. Johnson et al., Effect of neutrophil depletion on gelatinase expression, edema formation and hemorrhagic transformation after focal ischemic stroke, BMC Neurosci, vol.6, p.49, 2005.

K. Hayakawa, K. Mishima, M. Nozako, M. Hazekawa, S. Mishima et al., Delayed treatment with minocycline ameliorates neurologic impairment through activated microglia expressing a high-mobility group box1-inhibiting mechanism, Stroke, vol.39, pp.951-958, 2008.

T. Hayashi, S. Nagai, H. Fujii, Y. Baba, E. Ikeda et al., Critical roles of NK and CD8+ T cells in central nervous system listeriosis, J. Immunol, vol.182, pp.6360-6368, 2009.

J. He and F. T. Crews, Increased MCP-1 and microglia in various regions of the human alcoholic brain, Experimental Neurology, vol.210, pp.349-358, 2008.

E. Hennessy, É. W. Griffin, and C. Cunningham, Astrocytes Are Primed by Chronic Neurodegeneration to Produce Exaggerated Chemokine and Cell Infiltration Responses to Acute Stimulation with the Cytokines IL-? a d TNF-?, J. Neu os i, vol.35, pp.8411-8422, 2015.

J. Herz, P. Sabellek, T. E. Lane, M. Gunzer, D. M. Hermann et al., Role of Neutrophils in Exacerbation of Brain Injury After Focal Cerebral Ischemia in Hyperlipidemic Mice, Stroke, vol.46, pp.2916-2925, 2015.

W. F. Hickey and H. Kimura, Perivascular microglial cells of the CNS are bone marrow-derived and present antigen in vivo, Science, vol.239, pp.290-292, 1988.

G. E. Holder, C. M. Mcgary, E. M. Johnson, R. Zheng, V. T. John et al., Expression of the Mannose Receptor CD206 in HIV and SIV Encephalitis: A Phenotypic Switch of Brain Perivascular Macrophages with Virus Infection, Journal of Neuroimmune Pharmacology, vol.9, pp.716-726, 2014.

S. Homsi, T. Piaggio, N. Croci, F. Noble, M. Plotkine et al., Blockade of Acute Microglial Activation by Minocycline Promotes Neuroprotection and Reduces Locomotor Hyperactivity after Closed Head Injury in Mice: A Twelve-Week Follow-Up Study, Journal of Neurotrauma, vol.27, pp.911-921, 2010.

R. S. Hotchkiss, G. Monneret, and D. Payen, Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy, Nature Reviews Immunology, vol.13, pp.862-874, 2013.

M. M. Hughes, R. H. Field, V. H. Perry, C. L. Murray, and C. Cunningham, Microglia in the degenerating brain are capable of phagocytosis of beads and of apoptotic cells, but do not efficiently remove PrPSc, even upon LPS stimulation, Glia, vol.58, pp.2017-2030, 2010.

C. Iadecola, The Pathobiology of Vascular Dementia, Neuron, vol.80, pp.844-866, 2013.

C. Iadecola and J. Anrather, The immunology of stroke: from mechanisms to translation, Nat. Med, vol.17, pp.796-808, 2011.

J. J. Iliff, M. Wang, Y. Liao, B. A. Plogg, W. Peng et al., A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, 2012.

M. C. Janelsins, M. A. Mastrangelo, K. M. Park, K. L. Sudol, W. C. Narrow et al., Chronic Neuron-Specific Tumor Necrosis FactorAlpha Expression Enhances the Local Inflammatory Environment Ultimately Leading to Neuronal Death in 3xTg-AD Mice, Am J Pathol, vol.173, pp.1768-1782, 2008.

N. Jiang, M. Chopp, and S. Chahwala, Neutrophil inhibitory factor treatment of focal cerebral ischemia in the rat, Brain Res, vol.788, pp.25-34, 1998.

A. Y. Jin, U. I. Tuor, D. Rushforth, J. Kaur, R. N. Muller et al., Reduced blood brain barrier breakdown in P-selectin deficient mice following transient ischemic stroke: a future therapeutic target for treatment of stroke, BMC Neurosci, vol.11, p.12, 2010.

W. Jin, S. X. Shi, .. Li, Z. Li, M. Wood et al., Depletion of microglia exacerbates postischemic inflammation and brain injury, J Cereb Blood Flow Metab, vol.37, pp.2224-2236, 2017.

P. Kadlecová, R. Andel, R. Mikulík, E. P. Handing, and N. L. Pedersen, Alcohol consumption at midlife and risk of stroke during 43 years of follow-up: cohort and twin analyses, Stroke, vol.46, pp.627-633, 2015.

P. Kadlecová, R. Andel, R. Mikulík, E. P. Handing, and N. L. Pedersen, Alcohol consumption at midlife and risk of stroke during 43 years of follow-up: cohort and twin analyses, Stroke, vol.46, pp.627-633, 2015.

C. J. Kane and P. D. Drew, Inflammatory responses to alcohol in the CNS: nuclear receptors as potential therapeutics for alcohol-induced neuropathologies, J. Leukoc. Biol, vol.100, pp.951-959, 2016.

C. J. Kane, K. D. Phelan, J. C. Douglas, G. Wagoner, J. W. Johnson et al., Effects of Ethanol on Immune Response in the Brain: Region-Specific Changes in Adolescent Versus Adult Mice, Alcoholism: Clinical and Experimental Research, vol.38, pp.384-391, 2014.

H. Karatas, S. E. Erdener, Y. Gursoy-ozdemir, G. Gurer, F. Soylemezoglu et al.,

, Thrombotic distal middle cerebral artery occlusion produced by topical FeCl(3) application: a novel model suitable for intravital microscopy and thrombolysis studies, J. Cereb. Blood Flow Metab, vol.31, pp.1452-1460

U. W. Kaunzner, M. M. Miller, A. Gottfried-blackmore, J. Gal-toth, J. C. Felger et al., Accumulation of resident and peripheral dendritic cells in the aging CNS, Neurobiol. Aging, vol.33, pp.681-693, 2012.

T. Kawai, A. , and S. , The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors, Nature Immunology, vol.11, pp.373-384, 2010.

J. Keaney and M. Campbell, The dynamic blood-brain barrier, FEBS Journal, vol.282, pp.4067-4079, 2015.

I. Keskin, M. Y. Gunal, N. Ayturk, U. Kilic, M. Ozansoy et al., Dose-dependent neuroprotective effect of enoxaparin on cold-induced traumatic brain injury, Neural Regen Res, vol.12, pp.761-764, 2017.

K. Kisler, A. R. Nelson, S. V. Rege, A. Ramanathan, Y. Wang et al., Pericyte degeneration leads to neurovascular uncoupling and limits oxygen supply to brain, Nat. Neurosci, vol.20, pp.406-416, 2017.

T. O. Kleine, Cellular immune surveillance of central nervous system bypasses blood-brain barrier and blood-cerebrospinal-fluid barrier: Revealed with the New Marburg cerebrospinal-fluid model in healthy humans, Cytometry Part A, vol.87, pp.227-243, 2015.

C. Kleinschnitz, P. Kraft, A. Dreykluft, I. Hagedorn, K. Göbel et al., Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature, Blood, vol.121, pp.679-691, 2013.

E. Kohler, D. A. Prentice, T. R. Bates, G. J. Hankey, A. Claxton et al., Intravenous minocycline in acute stroke: a randomized, controlled pilot study and meta-analysis, Stroke, vol.44, pp.2493-2499, 2013.

S. A. Kolakowsky-hayner, E. V. Gourley, J. S. Kreutzer, J. H. Marwitz, D. X. Cifu et al.,

, Pre-injury substance abuse among persons with brain injury and persons with spinal cord injury, Brain Inj, vol.13, pp.571-581

A. D. Korczyn, Vascular parkinsonism-characteristics, pathogenesis and treatment, Nature Reviews Neurology, vol.11, pp.319-326, 2015.

N. Kostulas, H. Li, B. Xiao, Y. Huang, V. Kostulas et al., Dendritic cells are present in ischemic brain after permanent middle cerebral artery occlusion in the rat, Stroke, vol.33, pp.1129-1134, 2002.

E. Kovesdi, A. Kamnaksh, D. Wingo, F. Ahmed, N. E. Grunberg et al., Acute minocycline treatment mitigates the symptoms of mild blast-induced traumatic brain injury, Front Neurol, vol.3, p.111, 2012.

G. Krabbe, S. S. Minami, J. I. Etchegaray, P. Taneja, B. Djukic et al.,

, Mi oglial NF?B-TNF? h pe a ti atio i du es o sessi e-compulsive behavior in mouse models of progranulin-deficient frontotemporal dementia, 201700477.

T. Kristián and B. K. Siesjö, Calcium-related damage in ischemia, Life Sciences, vol.59, pp.357-367, 1996.

K. Kumasaka, J. A. Marks, R. Eisenstadt, M. A. Murcy, D. Samadi et al., In vivo leukocyte-mediated brain microcirculatory inflammation: a comparison of osmotherapies and progesterone in severe traumatic brain injury, Am. J. Surg, vol.208, pp.967-968, 2014.

V. Labat-gest and S. Tomasi, Photothrombotic ischemia: a minimally invasive and reproducible photochemical cortical lesion model for mouse stroke studies, J Vis Exp, p.289, 2013.

P. Lacy, Mechanisms of degranulation in neutrophils, Allergy Asthma Clin Immunol, vol.2, pp.98-108, 2006.

M. Lalancette-hébert, G. Gowing, A. Simard, Y. C. Weng, and J. Kriz, Selective ablation of proliferating microglial cells exacerbates ischemic injury in the brain, J. Neurosci, vol.27, pp.2596-2605, 2007.

Y. Lampl, M. Boaz, R. Gilad, M. Lorberboym, R. Dabby et al., Minocycline treatment in acute stroke: an open-label, evaluator-blinded study, Neurology, vol.69, pp.1404-1410, 2007.

J. A. Langlois, W. Rutland-brown, W. , and M. M. , The epidemiology and impact of traumatic brain injury: a brief overview, J Head Trauma Rehabil, vol.21, pp.375-378, 2006.

B. Lapergue, J. Labreuche, R. Blanc, X. Barreau, J. Berge et al., First-line use of contact aspiration for thrombectomy versus a stent retriever for recanalization in acute cerebral infarction: The randomized ASTER study protocol, Int J Stroke, vol.13, pp.87-95, 2018.

T. Lawrence, Coordinated Regulation of Signaling Pathways during Macrophage Activation, 2016.

T. Lawrence, D. A. Willoughby, and D. W. Gilroy, Anti-inflammatory lipid mediators and insights into the resolution of inflammation, Nat. Rev. Immunol, vol.2, pp.787-795, 2002.

L. J. Lawson, V. H. Perry, G. , and S. , Turnover of resident microglia in the normal adult mouse brain, Neuroscience, vol.48, pp.405-415, 1992.

L. Behot, A. Gauberti, M. Martinez-de-lizarrondo, S. Montagne, A. Lemarchand et al.,

, GpI ?-VWF blockade restores vessel patency by dissolving platelet aggregates formed under very high shear rate in mice, Blood, vol.123, pp.3354-3363

S. Leclercq, C. De-saeger, N. Delzenne, P. De-timary, and P. Stärkel, Role of Inflammatory Pathways, Blood Mononuclear Cells, and Gut-Derived Bacterial Products in Alcohol Dependence, Biological Psychiatry, vol.76, pp.725-733, 2014.

J. Lehmann, W. Härtig, A. Seidel, C. Füldner, C. Hobohm et al., Inflammatory cell recruitment after experimental thromboembolic stroke in rats, Neuroscience, vol.279, pp.139-154, 2014.

E. Lemarchand, M. Gauberti, S. Martinez-de-lizarrondo, H. Villain, Y. Repessé et al., Impact of alcohol consumption on the outcome of ischemic stroke and thrombolysis: role of the hepatic clearance of tissue-type plasminogen activator, Stroke, vol.46, pp.1641-1650, 2015.

Y. Li and M. Chopp, Temporal profile of nestin expression after focal cerebral ischemia in adult rat, Brain Research, vol.838, pp.1-10, 1999.

S. A. Liddelow, Fluids and barriers of the CNS: a historical viewpoint, Fluids Barriers CNS, vol.8, issue.2, 2011.

A. Liesz, E. Suri-payer, C. Veltkamp, H. Doerr, C. Sommer et al., , 2009.

, Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke, Nat. Med, vol.15, pp.192-199

A. Liesz, W. Zhou, É. Mracskó, S. Karcher, H. Bauer et al., Inhibition of lymphocyte trafficking shields the brain against deleterious neuroinflammation after stroke, Brain, vol.134, pp.704-720, 2011.

A. Liesz, W. Zhou, S. Na, G. J. Hämmerling, N. Garbi et al., Boosting regulatory T cells limits neuroinflammation in permanent cortical stroke, J. Neurosci, vol.33, pp.17350-17362, 2013.

J. W. Lin, J. T. Tsai, L. M. Lee, C. M. Lin, C. C. Hung et al., Effect of hyperbaric oxygen on patients with traumatic brain injury, Acta Neurochir. Suppl, vol.101, pp.145-149, 2008.

D. Lippai, S. Bala, T. Csak, E. A. Kurt-jones, and G. Szabo, Chronic Alcohol-Induced microRNA-155 Contributes to Neuroinflammation in a TLR4-Dependent Manner in Mice, PLoS ONE, vol.8, p.70945, 2013.

D. Lippai, S. Bala, J. Petrasek, T. Csak, I. Levin et al., Alcohol-induced IL-? i the ai is ediated NLRP /ASC, 2013.

, J. Leukoc. Biol, vol.94, pp.171-182

M. L. Lipton, E. Gellella, C. Lo, T. Gold, B. A. Ardekani et al., , 2008.

, Multifocal white matter ultrastructural abnormalities in mild traumatic brain injury with cognitive disability: a voxel-wise analysis of diffusion tensor imaging, J. Neurotrauma, vol.25, pp.1335-1342

C. Liu, C. Wu, Q. Yang, J. Gao, L. Li et al., Macrophages Mediate the Repair of Brain Vascular Rupture through Direct Physical Adhesion and Mechanical Traction, Immunity, vol.44, pp.1162-1176, 2016.

G. Liu, R. J. Middleton, C. R. Hatty, W. W. Kam, .. Chan et al., The 18 kDa Translocator Protein, Microglia and Neuroinflammation: TSPO, Microglia and Neuroinflammation, vol.24, pp.631-653, 2014.

Y. Liu, D. M. Jacobowitz, F. Barone, R. Mccarron, M. Spatz et al., Quantitation of Perivascular Monocytes and Macrophages around Cerebral Blood Vessels of Hypertensive and Aged Rats, Journal of Cerebral Blood Flow & Metabolism, vol.14, pp.348-352, 1994.

Z. Liu, Y. Li, Y. Cui, C. Roberts, M. Lu et al., Beneficial effects of gfap/vimentin reactive astrocytes for axonal remodeling and motor behavioral recovery in mice after stroke, Glia, vol.62, pp.2022-2033

G. Llovera, C. Benakis, G. Enzmann, R. Cai, T. Arzberger et al., The choroid plexus is a key cerebral invasion route for T cells after stroke, Acta Neuropathologica, vol.134, pp.851-868, 2017.

D. J. Loane and A. I. Faden, Neuroprotection for traumatic brain injury: translational challenges and emerging therapeutic strategies, Trends Pharmacol. Sci, vol.31, pp.596-604, 2010.

D. J. Loane, A. Kumar, B. A. Stoica, R. Cabatbat, and A. I. Faden, Progressive neurodegeneration after experimental brain trauma: association with chronic microglial activation, J. Neuropathol. Exp. Neurol, vol.73, pp.14-29, 2014.

D. Lobsien, M. Gawlitza, A. Schaudinn, S. Schob, C. Hobohm et al., Mechanical thrombectomy versus systemic thrombolysis in MCA stroke: a distance to thrombus-based outcome analysis, Journal of NeuroInterventional Surgery, vol.8, pp.878-882, 2016.

J. M. Loftis, M. Huckans, S. Ruimy, D. J. Hinrichs, and P. Hauser, Depressive symptoms in patients with chronic hepatitis C are correlated with elevated plasma levels of interleukin-1beta and tumor necrosis factor-alpha, Neurosci. Lett, vol.430, pp.264-268, 2008.

A. Louvet and P. Mathurin, Alcoholic liver disease: mechanisms of injury and targeted treatment, Nat Rev Gastroenterol Hepatol, vol.12, pp.231-242, 2015.

D. Lu, A. Goussev, J. Chen, P. Pannu, Y. Li et al., Atorvastatin reduces neurological deficit and increases synaptogenesis, angiogenesis, and neuronal survival in rats subjected to traumatic brain injury, J. Neurotrauma, vol.21, pp.21-32, 2004.

I. Lundgaard, W. Wang, A. Eberhardt, H. S. Vinitsky, B. C. Reeves et al., Beneficial effects of low alcohol exposure, but adverse effects of high alcohol intake on glymphatic function, 2018.

R. Macrez, C. Ali, O. Toutirais, B. Le-mauff, G. Defer et al., Stroke and the immune system: from pathophysiology to new therapeutic strategies, Lancet Neurol, vol.10, pp.471-480, 2011.

F. M. Marelli-be-g, M. Cle-e-t, C. Mau-o, and G. Caligiu-i, A i u ologist's guide to CD function in T-cells, J Cell Sci, vol.126, pp.2343-2352

S. Margulies and R. Hicks, Combination therapies for traumatic brain injury: prospective considerations, J. Neurotrauma, vol.26, pp.925-939, 2009.

A. Marmarou, M. A. Foda, W. Van-den-brink, J. Campbell, H. Kita et al., A new model of diffuse brain injury in rats. Part I: Pathophysiology and biomechanics, J. Neurosurg, vol.80, pp.291-300, 1994.

M. Mato, S. Ookawara, and K. Kurihara, Uptake of exogenous substances and marked infoldings of the fluorescent granular pericyte in cerebral fine vessels, Am. J. Anat, vol.157, pp.329-332, 1980.

N. Matsukawa, T. Yasuhara, K. Hara, L. Xu, M. Maki et al., Therapeutic targets and limits of minocycline neuroprotection in experimental ischemic stroke, BMC Neurosci, vol.10, p.126, 2009.

Y. Matsuo, H. Onodera, Y. Shiga, M. Nakamura, M. Ninomiya et al., , 1994.

, Correlation between myeloperoxidase-quantified neutrophil accumulation and ischemic brain injury in the rat. Effects of neutrophil depletion, Stroke, vol.25, pp.1469-1475

A. T. Mazzeo, O. L. Alves, C. B. Gilman, R. L. Hayes, C. Tolias et al., Brain metabolic and hemodynamic effects of cyclosporin A after human severe traumatic brain injury: a microdialysis study, Acta Neurochir, vol.150, pp.1019-1031, 2008.

T. K. Mcintosh, R. Vink, L. Noble, I. Yamakami, S. Fernyak et al., Traumatic brain injury in the rat: characterization of a lateral fluid-percussion model, Neuroscience, vol.28, pp.233-244, 1989.

P. G. Mcmenamin, Distribution and phenotype of dendritic cells and resident tissue macrophages in the dura mater, leptomeninges, and choroid plexus of the rat brain as demonstrated in wholemount preparations, J. Comp. Neurol, vol.405, pp.553-562, 1999.

A. J. Mierzwa, C. M. Marion, G. M. Sullivan, D. P. Mcdaniel, A. et al., Components of myelin damage and repair in the progression of white matter pathology after mild traumatic brain injury, J. Neuropathol. Exp. Neurol, vol.74, pp.218-232, 2015.

S. I. Miller, R. K. Ernst, and M. W. Bader, LPS, TLR4 and infectious disease diversity, Nature Reviews Microbiology, vol.3, pp.36-46, 2005.

E. A. Mistry, A. M. Mistry, M. O. Nakawah, R. V. Chitale, R. F. James et al., , 2017.

, Mechanical Thrombectomy Outcomes With and Without Intravenous Thrombolysis in Stroke Patients: A Meta-Analysis, Stroke, vol.48, pp.2450-2456

A. Montagne, M. Gauberti, R. Macrez, A. Jullienne, A. Briens et al., Ultra-sensitive molecular MRI of cerebrovascular cell activation enables early detection of chronic central nervous system disorders, NeuroImage, vol.63, pp.760-770, 2012.

N. Morelli, E. Rota, E. Michieletti, and D. Guidetti, Mechanical thrombectomy after intravenous thrombolysis for acute ischaemic stroke, The Lancet Neurology, vol.16, pp.103-104, 2017.

S. Mori and C. P. Leblond, Identification of microglia in light and electron microscopy, J. Comp. Neurol, vol.135, pp.57-80, 1969.

N. Morimoto, M. Shimazawa, T. Yamashima, H. Nagai, and H. Hara, Minocycline inhibits oxidative stress and decreases in vitro and in vivo ischemic neuronal damage, Brain Res, vol.1044, pp.8-15, 2005.

M. M. Muccigrosso, J. Ford, B. Benner, D. Moussa, C. Burnsides et al., Cognitive deficits develop 1month after diffuse brain injury and are exaggerated by microglia-associated reactivity to peripheral immune challenge, Brain Behav. Immun, vol.54, pp.95-109, 2016.

W. A. Muller, Leukocyte-endothelial cell interactions in the inflammatory response, Lab. Invest, vol.82, pp.521-533, 2002.

P. J. Murray, J. E. Allen, S. K. Biswas, E. A. Fisher, D. W. Gilroy et al., Macrophage activation and polarization: nomenclature and experimental guidelines, Immunity, vol.41, pp.14-20, 2014.

I. Napoli and H. Neumann, Microglial clearance function in health and disease, Neuroscience, vol.158, pp.1030-1038, 2009.

, Tissue plasminogen activator for acute ischemic stroke, N. Engl. J. Med, vol.333, pp.1581-1587, 1995.

J. T. Neary, Y. Kang, M. Tran, and J. Feld, Traumatic Injury Activates Protein Kinase B/Akt in Cultured Astrocytes: Role of Extracellular ATP and P2 Purinergic Receptors, Journal of Neurotrauma, vol.22, pp.491-500, 2005.

M. G. Netea, L. A. Jooste, E. Latz, K. H. Mills, G. Natoli et al., Trained immunity: A program of innate immune memory in health and disease, Science, vol.352, 1098.

J. Neumann, M. Riek-burchardt, J. Herz, T. R. Doeppner, R. König et al., Very-late-antigen-4 (VLA-4)-mediated brain invasion by neutrophils leads to interactions with microglia, increased ischemic injury and impaired behavior in experimental stroke, Acta Neuropathol, vol.129, pp.259-277, 2015.

S. N. Niogi, P. Mukherjee, J. Ghajar, C. Johnson, R. A. Kolster et al., Extent of microstructural white matter injury in postconcussive syndrome correlates with impaired cognitive reaction time: a 3T diffusion tensor imaging study of mild traumatic brain injury, AJNR Am J Neuroradiol, vol.29, pp.967-973, 2008.

D. M. Norden and J. P. Godbout, Review: microglia of the aged brain: primed to be activated and resistant to regulation, Neuropathol. Appl. Neurobiol, vol.39, pp.19-34, 2013.

D. M. Norden, M. M. Muccigrosso, and J. P. Godbout, Microglial priming and enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and neurodegenerative disease, Neuropharmacology, vol.96, pp.29-41, 2015.

M. J. O'do-ell, D. Xa-ie, L. Liu, H. Zha-g, S. L. Chi et al., Risk factors for ischaemic and intracerebral haemorrhagic stroke in 22 countries (the INTERSTROKE study): a case-control study, Lancet, vol.376, pp.112-123, 2010.

J. G. O'lea, M. Gooda-zi, D. L. Ia, and U. H. , T ell-and B cell-independent adaptive immunity mediated by natural killer cells, Nat. Immunol, vol.7, pp.507-516

G. C. Olivera, X. Ren, S. K. Vodnala, J. Lu, L. Coppo et al., Nitric Oxide Protects against Infection-Induced Neuroinflammation by Preserving the Stability of the Blood-Brain Barrier, PLOS Pathogens, vol.12, p.1005442, 2016.

G. Onofre, M. Kolácková, K. Jankovicová, and J. Krejsek, Scavenger receptor CD163 and its biological functions, Acta Medica, vol.52, pp.57-61, 2009.

C. Orset, R. Macrez, A. R. Young, D. Panthou, E. Angles-cano et al., Mouse model of in situ thromboembolic stroke and reperfusion, Stroke, vol.38, pp.2771-2778, 2007.

C. Orset, B. Haelewyn, S. M. Allan, S. Ansar, F. Campos et al., Efficacy of Alteplase in a Mouse Model of Acute Ischemic Stroke: A Retrospective Pooled Analysis, Stroke, vol.47, pp.1312-1318, 2016.

L. W. Ostrow, T. M. Suchyna, and F. Sachs, Stretch induced endothelin-1 secretion by adult rat astrocytes involves calcium influx via stretch-activated ion channels (SACs), Biochemical and Biophysical Research Communications, vol.410, pp.81-86, 2011.

R. D. Oude-engberink, E. L. Blezer, E. I. Hoff, S. M. Van-der-pol, A. Van-der-toorn et al., MRI of monocyte infiltration in an animal model of neuroinflammation using SPIOlabeled monocytes or free USPIO, J. Cereb. Blood Flow Metab, vol.28, pp.841-851, 2008.

P. Srivastava, M. V. Bhasin, A. Bhatia, R. Garg, A. Gaikwad et al., Efficacy of minocycline in acute ischemic stroke: a single-blinded, placebo-controlled trial, Neurol India, vol.60, pp.23-28, 2012.

L. Park, K. Uekawa, L. Garcia-bonilla, K. Koizumi, M. Murphy et al., Brain Perivascular Macrophages Initiate the Neurovascular Dysfunction of Alzhei e A? Peptides, Circ. Res, vol.121, pp.258-269, 2017.

J. Pedragosa, A. Salas-perdomo, M. Gallizioli, R. Cugota, F. Miró-mur et al., CNS-border associated macrophages respond to acute ischemic stroke attracting granulocytes and promoting vascular leakage, Acta Neuropathol Commun, vol.6, p.76, 2018.

C. Perego, S. Fumagalli, D. Simoni, and M. , Temporal pattern of expression and colocalization of microglia/macrophage phenotype markers following brain ischemic injury in mice, J Neuroinflammation, vol.8, p.174, 2011.

I. Perez-de-puig, F. Miró-mur, M. Ferrer-ferrer, E. Gelpi, J. Pedragosa et al., Neutrophil recruitment to the brain in mouse and human ischemic stroke, Acta Neuropathol, vol.129, pp.239-257, 2015.

V. H. Perry and C. Holmes, Microglial priming in neurodegenerative disease, Nature Reviews Neurology, vol.10, pp.217-224, 2014.

V. H. Perry, J. A. Nicoll, and C. Holmes, Microglia in neurodegenerative disease, Nature Reviews Neurology, vol.6, pp.193-201, 2010.

L. Peruzzotti-jametti, M. Donegá, E. Giusto, G. Mallucci, B. Marchetti et al., The role of the immune system in central nervous system plasticity after acute injury, Neuroscience, vol.283, pp.210-221, 2014.

M. Pham, C. Kleinschnitz, X. Helluy, A. J. Bartsch, M. Austinat et al., Enhanced cortical reperfusion protects coagulation factor XII-deficient mice from ischemic stroke as revealed by high-field MRI, Neuroimage, vol.49, pp.2907-2914, 2010.

A. Pisanu, D. Lecca, G. Mulas, J. Wardas, G. Simbula et al., Dynamic changes in pro-and anti-inflammatory cytokines in microglia after PPAR-? ago ist eu op ote ti e treatment in the MPTPp ouse odel of p og essi e Pa ki so 's disease, Neu o iol. Dis, vol.71, pp.280-291, 2014.

B. A. Plog, M. L. Dashnaw, E. Hitomi, W. Peng, Y. Liao et al., Biomarkers of Traumatic Injury Are Transported from Brain to Blood via the Glymphatic System, Journal of Neuroscience, vol.35, pp.518-526, 2015.

M. M. Polfliet, P. H. Goede, E. M. Van-kesteren-hendrikx, N. Van-rooijen, C. D. Dijkstra et al., A method for the selective depletion of perivascular and meningeal macrophages in the central nervous system, J. Neuroimmunol, vol.116, pp.188-195, 2001.

M. M. Polfliet, F. Van-de-veerdonk, E. A. Döpp, E. M. Van-kesteren-hendrikx, N. Van-rooijen et al., The role of perivascular and meningeal macrophages in experimental allergic encephalomyelitis, J. Neuroimmunol, vol.122, pp.1-8, 2002.

B. Pradier, E. Erxlebe, A. Markert, and I. Rácz, Microglial IL-? p og essi el i eases ith duration of alcohol consumption, Naunyn Schmiedebergs Arch. Pharmacol, vol.391, pp.455-461, 2018.

A. Prakash, S. V. Parelkar, S. N. Oak, R. K. Gupta, B. V. Sanghvi et al., Role of hyperbaric oxygen therapy in severe head injury in children, J Pediatr Neurosci, vol.7, pp.4-8, 2012.

C. J. Prestigiacomo, S. C. Kim, E. S. Connolly, H. Liao, S. Yan et al., CD18-mediated neutrophil recruitment contributes to the pathogenesis of reperfused but not nonreperfused stroke, Stroke, vol.30, pp.1110-1117, 1999.

C. J. Price, D. Wang, D. K. Menon, J. V. Guadagno, M. Cleij et al., Intrinsic activated microglia map to the peri-infarct zone in the subacute phase of ischemic stroke, Stroke, vol.37, pp.1749-1753, 2006.

M. Prinz, D. Erny, and N. Hagemeyer, Ontogeny and homeostasis of CNS myeloid cells, Nat. Immunol, vol.18, pp.385-392, 2017.

P. Proost, A. Wuyts, and J. Van-damme, The role of chemokines in inflammation, Int. J. Clin. Lab. Res, vol.26, pp.211-223, 1996.

L. Qin, X. Wu, M. L. Block, Y. Liu, G. R. Breese et al., Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration, Glia, vol.55, pp.453-462, 2007.

J. Quintin, S. Saeed, J. H. Martens, E. J. Giamarellos-bourboulis, D. C. Ifrim et al., Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes, Cell Host Microbe, vol.12, pp.223-232, 2012.

V. Ramaglia, T. R. Hughes, R. M. Donev, M. M. Ruseva, X. Wu et al., C3-dependent mechanism of microglial priming relevant to multiple sclerosis, Proc. Natl. Acad. Sci. U.S.A, vol.109, pp.965-970, 2012.

R. Raychev and J. L. Saver, Mechanical thrombectomy devices for treatment of stroke, Neurol Clin Pract, vol.2, pp.231-235, 2012.

D. V. Reneer, R. D. Hisel, J. M. Hoffman, R. J. Kryscio, B. T. Lusk et al., A multi-mode shock tube for investigation of blast-induced traumatic brain injury, J. Neurotrauma, vol.28, pp.95-104, 2011.

R. Reshef, E. Kudryavitskaya, H. Shani-narkiss, B. Isaacson, N. Rimmerman et al., The role of microglia and their CX3CR1 signaling in adult neurogenesis in the olfactory bulb, 2017.

R. A. Rice, E. E. Spangenberg, H. Yamate-morgan, R. J. Lee, R. P. Arora et al., Elimination of Microglia Improves Functional Outcomes Following Extensive Neuronal Loss in the Hippocampus, J. Neurosci, vol.35, pp.9977-9989, 2015.

D. Ricklin, D. C. Mastellos, E. S. Reis, and J. D. Lambris, The renaissance of complement therapeutics, Nat Rev Nephrol, vol.14, pp.26-47, 2018.

M. Rocha-e-silva, A brief survey of the history of inflammation, Agents Actions, vol.8, pp.45-49, 1978.

G. L. Rockswold, S. E. Ford, D. C. Anderson, T. A. Bergman, and R. E. Sherman, Results of a prospective randomized trial for treatment of severely brain-injured patients with hyperbaric oxygen, J. Neurosurg, vol.76, pp.929-934, 1992.

S. B. Rockswold, G. L. Rockswold, J. M. Vargo, C. A. Erickson, R. L. Sutton et al., Effects of hyperbaric oxygenation therapy on cerebral metabolism and intracranial pressure in severely brain injured patients, J. Neurosurg, vol.94, pp.403-411, 2001.

S. B. Rockswold, G. L. Rockswold, D. A. Zaun, X. Zhang, C. E. Cerra et al., A prospective, randomized clinical trial to compare the effect of hyperbaric to normobaric hyperoxia on cerebral metabolism, intracranial pressure, and oxygen toxicity in severe traumatic brain injury, J. Neurosurg, vol.112, pp.1080-1094, 2010.

S. B. Rockswold, G. L. Rockswold, D. A. Zaun, and J. Liu, A prospective, randomized Phase II clinical trial to evaluate the effect of combined hyperbaric and normobaric hyperoxia on cerebral metabolism, intracranial pressure, oxygen toxicity, and clinical outcome in severe traumatic brain injury, J. Neurosurg, vol.118, pp.1317-1328, 2013.

J. Rodhe, M. A. Burguillos, R. M. De-pablos, E. Kavanagh, A. Persson et al., Spatio-temporal activation of caspase-8 in myeloid cells upon ischemic stroke, Acta Neuropathologica Communications, vol.4, p.92, 2016.

L. Romani, Immunity to fungal infections, Nat. Rev. Immunol, vol.4, pp.1-23, 2004.

V. Rubovitch, M. Ten-bosch, O. Zohar, C. R. Harrison, C. Tempel-brami et al., A mouse model of blast-induced mild traumatic brain injury, Exp. Neurol, vol.232, pp.280-289, 2011.

R. L. Sacco, M. Elkind, B. Boden-albala, I. F. Lin, D. E. Kargman et al., The protective effect of moderate alcohol consumption on ischemic stroke, JAMA, vol.281, pp.53-60, 1999.

S. Saeed, J. Quintin, H. H. Kerstens, N. A. Rao, A. Aghajanirefah et al., Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity, Science, vol.345, p.1251086, 2014.

K. Saijo and C. K. Glass, Microglial cell origin and phenotypes in health and disease, Nature Reviews Immunology, vol.11, pp.775-787, 2011.

E. Santos, M. Schöll, R. Sánchez-porras, M. A. Dahlem, H. Silos et al., Radial, spiral and reverberating waves of spreading depolarization occur in the gyrencephalic brain, NeuroImage, vol.99, pp.244-255, 2014.

N. R. Saunders, J. Dreifuss, K. M. Dziegielewska, P. A. Johansson, M. D. Habgood et al., The rights and wrongs of blood-brain barrier permeability studies: a walk through 100 years of history, Front Neurosci, vol.8, p.404, 2014.

M. Schilling, M. Besselmann, M. Müller, J. K. Strecker, E. B. Ringelstein et al., , 2005.

, 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, vol.196, pp.290-297

A. Schmidt, J. Strecker, S. Hucke, N. Bruckmann, M. Herold et al., Targeting Different Monocyte/Macrophage Subsets Has No Impact on Outcome in Experimental Stroke, Stroke, vol.48, pp.1061-1069, 2017.

J. W. Schouten, Neuroprotection in traumatic brain injury: a complex struggle against the biology of nature, Curr Opin Crit Care, vol.13, pp.134-142, 2007.

G. Scott, H. Zetterberg, A. Jolly, J. H. Cole, S. De-simoni et al., Minocycline reduces chronic microglial activation after brain trauma but increases neurodegeneration, Brain, vol.141, pp.459-471, 2018.

R. Sharma and D. T. Laskowitz, Biomarkers in traumatic brain injury, Curr Neurol Neurosci Rep, vol.12, pp.560-569, 2012.

D. A. Shear, X. M. Lu, M. C. Bombard, R. Pedersen, Z. Chen et al., Longitudinal characterization of motor and cognitive deficits in a model of penetrating ballistic-like brain injury, J. Neurotrauma, vol.27, pp.1911-1923, 2010.

D. A. Shear, X. M. Lu, R. Pedersen, G. Wei, Z. Chen et al., Severity profile of penetrating ballistic-like brain injury on neurofunctional outcome, blood-brain barrier permeability, and brain edema formation, J. Neurotrauma, vol.28, pp.2185-2195, 2011.

D. Shi, J. Zhang, H. Jiang, C. Tong, G. Gu et al., LPS pretreatment ameliorates multiple organ injuries and improves survival in a murine model of polymicrobial sepsis, Inflamm. Res, vol.60, pp.841-849, 2011.

J. E. Siegler, A. K. Boehme, A. D. Kumar, M. A. Gillette, K. C. Albright et al., Identification of modifiable and non-modifiable risk factors for neurological deterioration following acute ischemic stroke, J Stroke Cerebrovasc Dis, vol.22, pp.207-213, 2013.

J. Silver and J. H. Miller, Regeneration beyond the glial scar, Nat. Rev. Neurosci, vol.5, pp.146-156, 2004.
DOI : 10.1038/nrn1326

URL : https://www.nature.com/articles/nrn1326.pdf

N. Simon, K. Buschmann, J. Pflaum, H. Hudalla, L. Koch et al.,

, Anti-Inflammatory Functions of Protein C Require RAGE and ICAM-1 in a Stimulus-Dependent Manner, Mediators of Inflammation, p.743678, 2014.

A. K. Singh, Y. Jiang, S. Gupta, and E. Benlhabib, Effects of chronic ethanol drinking on the blood brain barrier and ensuing neuronal toxicity in alcohol-preferring rats subjected to intraperitoneal LPS injection, Alcohol Alcohol, vol.42, pp.385-399, 2007.

B. E. Skolnick, A. I. Maas, R. K. Narayan, R. G. Van-der-hoop, T. Macallister et al., A clinical trial of progesterone for severe traumatic brain injury, N. Engl. J. Med, vol.371, pp.2467-2476, 2014.

L. M. Sly, R. F. Krzesicki, J. R. Brashler, A. E. Buhl, D. D. Mckinley et al., , 2001.

, Endogenous brain cytokine mRNA and inflammatory responses to lipopolysaccharide are elevated in the Tg t a sge i ouse odel of Alzhei e 's disease, B ai Res. Bull, vol.56, pp.581-588

C. J. Smith, S. Hulme, A. Vail, C. Heal, A. R. Parry-jones et al., SCIL-STROKE (Subcutaneous Interleukin-1 Receptor Antagonist in Ischemic Stroke): A, 2018.

C. L. Sokol and A. D. Luster, The Chemokine System in Innate Immunity, Cold Spring Harb Perspect Biol, 2015.

C. Soulas, R. E. Donahue, C. E. Dunbar, D. A. Persons, X. Alvarez et al., Genetically Modified CD34+ Hematopoietic Stem Cells Contribute to Turnover of Brain Perivascular Macrophages in Long-Term Repopulated Primates, The American Journal of Pathology, vol.174, pp.1808-1817, 2009.

P. G. Sullivan, M. Thompson, and S. W. Scheff, Continuous infusion of cyclosporin A postinjury significantly ameliorates cortical damage following traumatic brain injury, Exp. Neurol, vol.161, pp.631-637, 2000.

M. D. Sweeney, A. P. Sagare, and B. V. Zlokovic, Cerebrospinal Fluid Biomarkers of Neurovascular D sfu tio i Mild De e tia a d Alzhei e 'S Disease, vol.35, pp.1055-1068, 2015.

G. Szabo, Gut-Liver Axis in Alcoholic Liver Disease, Gastroenterology, vol.148, pp.30-36, 2015.

G. Szalay, B. Martinecz, N. Lénárt, Z. Környei, B. Orsolits et al., Microglia protect against brain injury and their selective elimination dysregulates neuronal network activity after stroke, Nature Communications, vol.7, p.11499, 2016.

J. Szmydynger-chodobska, N. Strazielle, J. R. Gandy, T. H. Keefe, B. J. Zink et al., Posttraumatic invasion of monocytes across the blood-cerebrospinal fluid barrier, J. Cereb. Blood Flow Metab, vol.32, pp.93-104, 2012.

N. Tajiri, S. L. Kellogg, T. Shimizu, G. W. Arendash, and C. V. Borlongan, Traumatic brain injury, 2013.

J. H. Tapia-perez, M. Sanchez-aguilar, J. G. Torres-corzo, A. Gordillo-moscoso, P. Martinez-perez et al., Effect of rosuvastatin on amnesia and disorientation after traumatic brain injury (NCT003229758), J. Neurotrauma, vol.25, pp.1011-1017, 2008.

A. Taylor, J. Verhagen, K. Blaser, M. Akdis, and C. A. Akdis, Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: the role of T regulatory cells, Immunology, vol.117, pp.433-442, 2006.

A. Thiel and W. Heiss, Imaging of Microglia Activation in Stroke, Stroke, vol.42, pp.507-512, 2011.

R. D. Thompson, K. E. Noble, K. Y. Larbi, A. Dewar, G. S. Duncan et al., , 2001.

, Platelet-endothelial cell adhesion molecule-1 (PECAM-1)-deficient mice demonstrate a transient and cytokine-specific role for PECAM-1 in leukocyte migration through the perivascular basement membrane, Blood, vol.97, pp.1854-1860

C. L. Tinianow, T. K. Tinianow, and M. Wilcox, Effects of hyperbaric oxygen on focal brain contusions, Biomed Sci Instrum, vol.36, pp.275-281, 2000.

Q. Tuo, P. Lei, K. Jackman, X. Li, H. Xiong et al., Tau-mediated iron export prevents ferroptotic damage after ischemic stroke, Molecular Psychiatry, vol.22, 2017.

M. D. Turner, B. Nedjai, T. Hurst, and D. J. Pennington, Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease, Biochimica et Biophysica Acta (BBA) -Molecular Cell Research, vol.1843, pp.2563-2582, 2014.

N. H. Varvel, J. J. Neher, A. Bosch, W. Wang, R. M. Ransohoff et al., Infiltrating monocytes promote brain inflammation and exacerbate neuronal damage after status epilepticus, Proc. Natl. Acad. Sci. U.S.A, vol.113, pp.5665-5674, 2016.

D. Vestweber, How leukocytes cross the vascular endothelium, Nature Reviews Immunology, vol.15, pp.692-704, 2015.

S. Vidale, M. Longoni, L. Valvassori, A. , and E. , Mechanical Thrombectomy in Strokes with Large-Vessel Occlusion Beyond 6 Hours: A Pooled Analysis of Randomized Trials, J Clin Neurol, vol.14, pp.407-412, 2018.

V. Bernhardi, R. Eugenin-von, L. Bernhardi, and J. Eugenin, Microglial cell dysregulation in Brain Aging and Neurodegeneration. Front. Aging Neurosci, vol.7, 2015.

H. J. Wang, Alcohol, inflammation, and gut-liver-brain interactions in tissue damage and disease development, World Journal of Gastroenterology, vol.16, 1304.

C. X. Wang, T. Yang, R. Noor, and A. Shuaib, Delayed minocycline but not delayed mild hypothermia protects against embolic stroke, BMC Neurol, vol.2, issue.2, 2002.

G. Wang, J. Zhang, X. Hu, L. Zhang, L. Mao et al.,

, Microglia/macrophage polarization dynamics in white matter after traumatic brain injury, J. Cereb. Blood Flow Metab, vol.33, pp.1864-1874

Y. F. Wang, S. E. Tsirka, S. Strickland, P. E. Stieg, S. G. Soriano et al., Tissue plasminogen activator (tPA) increases neuronal damage after focal cerebral ischemia in wild-type and tPA-deficient mice, Nat. Med, vol.4, pp.228-231, 1998.

I. B. Wanner, M. A. Anderson, B. Song, J. Levine, A. Fernandez et al., Glial Scar Borders Are Formed by Newly Proliferated, Elongated Astrocytes That Interact to Corral Inflammatory and Fibrotic Cells via STAT3-Dependent Mechanisms after Spinal Cord Injury, J. Neurosci, vol.33, pp.12870-12886, 2013.

S. Wattananit, D. Tornero, N. Graubardt, T. Memanishvili, E. Monni et al., Monocyte-Derived Macrophages Contribute to Spontaneous Long-Term Functional Recovery after Stroke in Mice, J. Neurosci, vol.36, pp.4182-4195, 2016.

J. Wei, X. , and G. , The neuroprotective effects of progesterone on traumatic brain injury: current status and future prospects, Acta Pharmacol. Sin, vol.34, pp.1485-1490, 2013.

J. R. Weinstein, I. P. Koerner, and T. Möller, Microglia in ischemic brain injury, Future Neurol, vol.5, pp.227-246, 2010.

C. Werner and K. Engelhard, Pathophysiology of traumatic brain injury, Br J Anaesth, vol.99, pp.4-9, 2007.

S. Wernersson and G. Pejler, Mast cell secretory granules: armed for battle, Nat. Rev. Immunol, vol.14, pp.478-494, 2014.

L. Whitnall, T. M. Mcmillan, G. D. Murray, and G. M. Teasdale, Disability in young people and adults after head injury: 5-7 year follow up of a prospective cohort study, J. Neurol. Neurosurg. Psychiatry, vol.77, pp.640-645, 2006.

E. A. Winkler, J. D. Sengillo, J. S. Sullivan, J. S. Henkel, S. H. Appel et al., Blood-spinal cord barrier breakdown and pericyte reductions in amyotrophic lateral sclerosis, Acta Neuropathol, vol.125, pp.111-120, 2013.

D. W. Wright, A. L. Kellermann, V. S. Hertzberg, P. L. Clark, M. Frankel et al., ProTECT: a randomized clinical trial of progesterone for acute traumatic brain injury, Ann Emerg Med, vol.49, pp.1-2, 2007.

D. W. Wright, S. D. Yeatts, R. Silbergleit, Y. Y. Palesch, V. S. Hertzberg et al., Very early administration of progesterone for acute traumatic brain injury, N. Engl. J. Med, vol.371, pp.2457-2466, 2014.

G. Xiao, J. Wei, W. Yan, W. Wang, and Z. Lu, Improved outcomes from the administration of progesterone for patients with acute severe traumatic brain injury: a randomized controlled trial, Crit Care, vol.12, p.61, 2008.

Y. Xiong, A. Mahmood, and M. Chopp, Animal models of traumatic brain injury, Nat. Rev. Neurosci, vol.14, pp.128-142, 2013.

L. Xu, S. C. Fagan, J. L. Waller, D. Edwards, C. V. Borlongan et al., Low dose intravenous minocycline is neuroprotective after middle cerebral artery occlusion-reperfusion in rats, BMC Neurol, vol.4, p.7, 2004.

J. Xue, S. V. Schmidt, J. Sander, A. Draffehn, W. Krebs et al., Transcriptome-based network analysis reveals a spectrum model of human macrophage activation, Immunity, vol.40, pp.274-288, 2014.

A. W. Yan, D. E. Fouts, J. Brandl, P. Stärkel, M. Torralba et al., Enteric dysbiosis associated with a mouse model of alcoholic liver disease, Hepatology, vol.53, pp.96-105, 2011.

J. Yang, T. Hirata, K. Croce, G. Merrill-skoloff, B. Tchernychev et al.,

B. Furie, Targeted Gene Disruption Demonstrates That P-Selectin Glycoprotein Ligand 1 (Psgl-1) Is Required for P-Selectin-Mediated but Not E-Selectin-Mediated Neutrophil Rolling and Migration, The Journal of Experimental Medicine, vol.190, pp.1769-1782, 2000.

Y. Yang, V. M. Salayandia, J. F. Thompson, L. Y. Yang, E. Y. Estrada et al., Attenuation of acute stroke injury in rat brain by minocycline promotes blood-brain barrier remodeling and alternative microglia/macrophage activation during recovery, Journal of Neuroinflammation, vol.12, p.26, 2015.

Y. Yang, H. Liu, H. Zhang, Q. Ye, J. Wang et al., , 2017.

, ST2/IL-33-Dependent Microglial Response Limits Acute Ischemic Brain Injury, J. Neurosci, vol.37, pp.4692-4704

Y. Yu, Z. Zhang, S. Wei, J. Serrats, R. M. Weiss et al., Brain Perivascular Macrophages and the Sympathetic Response to Inflammation in Rats After Myocardial Infarction, Hypertension, vol.55, pp.652-659, 2010.

H. Zetterberg and K. Blennow, Fluid biomarkers for mild traumatic brain injury and related conditions, Nature Reviews Neurology, vol.12, pp.563-574, 2016.

Z. Zhang, Z. Zhang, Y. Wu, and H. J. Schluesener, Lesional accumulation of CD163+ macrophages/microglia in rat traumatic brain injury, Brain Res, vol.1461, pp.102-110, 2012.

Z. Zhao, A. R. Nelson, C. Betsholtz, and B. V. Zlokovic, Establishment and Dysfunction of the Blood-Brain Barrier, Cell, vol.163, pp.1064-1078, 2015.

W. Zhou, A. Liesz, H. Bauer, C. Sommer, B. Lahrmann et al., , 2013.

, Postischemic brain infiltration of leukocyte subpopulations differs among murine permanent and transient focal cerebral ischemia models, Brain Pathol, vol.23, pp.34-44

X. Zhu, M. Fréchou, P. Liere, S. Zhang, A. Pianos et al., A Role of Endogenous Progesterone in Stroke Cerebroprotection Revealed by the Neural-Specific Deletion of Its Intracellular Receptors, The Journal of Neuroscience, vol.37, pp.10998-11020, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01623387

Z. Zhu, Y. Fu, D. Tian, N. Sun, W. Han et al., IL-33 attenuates EAE by suppressing IL-17 and IFN-gamma production and inducing alternatively activated macrophages, Combination of the Immune Modulator Fingolimod With Alteplase in Acute Ischemic Stroke: A Pilot Trial, vol.132, pp.9614-9616, 1992.

M. Pekny, M. Nilsson, C. Perego, S. Fumagalli, D. Simoni et al., Temporal pattern of expression and colocalization of microglia/macrophage phenotype markers following brain ischemic injury in mice, Astrocyte activation and reactive gliosis, vol.50, pp.11281-11291, 2005.

?. J. Neurosci, , vol.37, pp.9614-9616, 2017.

. Drieu, Journal Club

E. C. Jauch, J. L. Saver, H. P. Adams, A. Bruno, J. Connors et al., Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/ American Stroke Association, Stroke, vol.44, issue.3, pp.870-947, 2013.

T. Gaberel, C. Gakuba, M. Hebert, A. Montagne, V. Agin et al.,

, * -weighted images during normobaric oxygen therapy, Stroke, vol.44, issue.12, pp.3482-3491, 2013.

V. Rohde, I. Rohde, R. Thiex, A. Ince, A. Jung et al., Fibrinolysis therapy achieved with tissue plasminogen activator and aspiration of the liquefied clot after experimental intracerebral hemorrhage: rapid reduction in hematoma volume but intensification of delayed edema formation, J Neurosurg, vol.97, issue.4, pp.954-62, 2002.

C. Rorden, H. Karnath, and L. Bonilha, Improving lesion-symptom mapping, J Cogn Neurosci, vol.19, issue.7, pp.1081-1089, 2007.

K. R. Thulborn, J. C. Waterton, P. M. Matthews, and G. K. Radda, Oxygenation dependence of the transverse relaxation time of water protons in whole blood at high field, Biochim Biophys Acta, vol.714, issue.2, pp.265-70, 1982.

U. Dirnagl, Bench to bedside: the quest for quality in experimental stroke research, J Cereb Blood Flow Metab, vol.26, issue.12, pp.1465-78, 2006.

T. L. Sudduth, D. K. Powell, C. D. Smith, A. Greenstein, and D. M. Wilcock, Induction of hyperhomocysteinemia models vascular dementia by induction of cerebral microhemorrhages and neuroinflammation, J Cereb Blood Flow Metab, vol.33, issue.5, pp.708-723, 2013.

N. Fujiwara, E. T. Mandeville, X. Geng, Y. Luo, K. Arai et al., Effect of normobaric oxygen therapy in a rat model of intracerebral hemorrhage, Stroke, vol.42, issue.5, pp.1469-72, 2011.

I. Linfante, R. H. Llinas, L. R. Caplan, and S. Warach, MRI features of intracerebral hemorrhage within 2 hours from symptom onset, Stroke, vol.30, issue.11, pp.2263-2270, 1999.

R. Goulay, Journal of Clinical Neuroscience, 2018.

R. Goulay, Modification of apparent intracerebral hematoma volume on T2 ? -weighted images during normobaric oxygen therapy may contribute to false diagnosis, J Clin Neurosci, 2018.

N. Univ, I. Unicaen, and U. U1237, Physiopathology and Imaging of Neurological Disorders (PhIND)

I. Langevin and . Paris, CNRS UMR 7587, INSERM U979

, * Corresponding Author Information

V. Denis, I. Phd, and . Umr-s-u1237, Bd Becquerel, BP5229, 14074 Caen, France. Phone, fax and e-mail: +33 2 31 47 01 66, +33 2 31 47 02 22, vivien@cyceron.fr 1467, Santa-Cruz) primary antibodies, FITC-conjugated mouse anti-?SMA (1:500, ab8211, Abcam) primary antibodies and rabbit anti-laminin (1:2000, ab11575, Abcam) primary antibodies. Primary antibodies were revealed using Fab'2 fragment anti rabbit and goat IgG linked to CY3 and Alexa Fluor 647 (1:600, Jackson ImmunoResearch) co-incubated 90 min at room temperature. Vessels were then coverslipped using mounting medium containing DAPI, GIP Cyceron

, Fluorescent intensity of the GluN1 staining in vessels were assessed using ImageJ software (NIH)

, Arginine content in Actilyse was removed by dialysis at 4°C overnight (see rtPA preparation section). Then, purified rtPA was mixed with the N-succinimidyl ester of Alexa 555 for 4 h at 4°C with continuous stirring. The resulting solution was dialyzed in bicarbonate buffer overnight at 4°C to remove unbound dyes

, Cryomicrotome-cut sections (10 ?m) were collected on polylysine slides and stored at -80°C before processing. Sections were incubated overnight with FITC-conjugated mouse anti-?SMA (1:500, ab8211, Abcam) primary antibodies. Sections were then coverslipped using mounting medium containing DAPI

D. V. , A. A. , C. O. , A. D. , V. H. et al., Analyzing data: A.A and V.H. Writing the manuscript

C. Iadecola, The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease, Neuron, vol.96, pp.17-42, 2017.

D. Attwell, Glial and neuronal control of brain blood flow, Nature, vol.468, pp.232-243, 2010.

Y. Otsu, Calcium dynamics in astrocyte processes during neurovascular coupling, Nat. Neurosci, vol.18, pp.210-218, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01362700

K. Kisler, Pericyte degeneration leads to neurovascular uncoupling and limits oxygen supply to brain, Nat. Neurosci, vol.20, pp.406-416, 2017.

B. R. Chen, M. G. Kozberg, M. B. Bouchard, M. A. Shaik, and E. M. Hillman, A Critical Role for the Vascular Endothelium in Functional Neurovascular Coupling in the Brain, J. Am. Heart Assoc, vol.3, p.787, 2014.

R. A. Hill, Regional Blood Flow in the Normal and Ischemic Brain Is Controlled by Arteriolar Smooth Muscle Cell Contractility and Not by Capillary Pericytes, Neuron, vol.87, pp.95-110, 2015.

C. N. Hall, Capillary pericytes regulate cerebral blood flow in health and disease, Nature, vol.508, pp.55-60, 2014.

F. Fernández-klett, N. Offenhauser, U. Dirnagl, J. Priller, and U. Lindauer, Pericytes in capillaries are contractile in vivo, but arterioles mediate functional hyperemia in the mouse brain, Proc. Natl. Acad. Sci, vol.107, pp.22290-22295, 2010.

D. Collen and H. R. Lijnen, Basic and clinical aspects of fibrinolysis and thrombolysis, Blood, vol.78, pp.3114-3124, 1991.

D. Vivien, M. Gauberti, A. Montagne, G. Defer, and E. Touzé, Impact of tissue plasminogen activator on the neurovascular unit: from clinical data to experimental evidence, J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab, vol.31, pp.2119-2134, 2011.

M. J. Alberts, T. Shang, and A. Magadan, Endovascular Therapy for Acute Ischemic Stroke: Dawn of a New Era, JAMA Neurol, vol.72, pp.1101-1103, 2015.

A. L. Samson and R. L. Medcalf, Tissue-type plasminogen activator: a multifaceted modulator of neurotransmission and synaptic plasticity, Neuron, vol.50, pp.673-678, 2006.

O. Nicole, The proteolytic activity of tissue-plasminogen activator enhances NMDARmediated signaling, Nat. Med, vol.7, pp.59-64, 2001.

F. Cassé, Glutamate controls tPA recycling by astrocytes, which in turn influences glutamatergic signals, J. Neurosci. Off. J. Soc. Neurosci, vol.32, pp.5186-5199, 2012.

R. Pawlak, Rapid, specific and active site-catalyzed effect of tissue-plasminogen activator on hippocampus-dependent learning in mice, Neuroscience, vol.113, pp.995-1001, 2002.

R. Madani, Enhanced hippocampal long-term potentiation and learning by increased neuronal expression of tissue-type plasminogen activator in transgenic mice, EMBO J, vol.18, pp.3007-3012, 1999.

R. Pawlak, A. M. Magarinos, J. Melchor, B. Mcewen, and S. Strickland, Tissue plasminogen activator in the amygdala is critical for stress-induced anxiety-like behavior, Nat. Neurosci, vol.6, pp.168-174, 2003.

T. Matys, Tissue plasminogen activator promotes the effects of corticotropin-releasing factor on the amygdala and anxiety-like behavior, Proc. Natl. Acad. Sci. U. S. A, vol.101, pp.16345-16350, 2004.

A. Chevilley, Impacts of tissue-type plasminogen activator (tPA) on neuronal survival, Front. Cell. Neurosci, vol.9, 2015.

J. Parcq, Unveiling an exceptional zymogen: the single-chain form of tPA is a selective activator of NMDAR-dependent signaling and neurotoxicity, Cell Death Differ, vol.19, 1983.

M. Hébert, Distant Space Processing is Controlled by tPA-dependent NMDAR Signaling in the Entorhinal Cortex, Cereb. Cortex, vol.27, pp.4783-4796, 2017.

L. Park, Key role of tissue plasminogen activator in neurovascular coupling, Proc. Natl. Acad. Sci. 105, pp.1073-1078, 2008.

J. E. Lochner, Activity-dependent release of tissue plasminogen activator from the dendritic spines of hippocampal neurons revealed by live-cell imaging, J. Neurobiol, vol.66, pp.564-577, 2006.

E. G. Levin and D. J. Loskutoff, Cultured bovine endothelial cells produce both urokinase and tissue-type plasminogen activators, J. Cell Biol, vol.94, pp.631-636, 1982.

E. Angles-cano, Production of monoclonal antibodies to the high fibrin-affinity, tissuetype plasminogen activator of human plasma. Demonstration of its endothelial origin by immunolocalization, Blood, vol.66, pp.913-920, 1985.

I. E. András, The NMDA and AMPA/KA Receptors are Involved in Glutamate-Induced Alterations of Occludin Expression and Phosphorylation in Brain Endothelial Cells, J. Cereb. Blood Flow Metab, vol.27, pp.1431-1443, 2007.

A. Reijerkerk, The NR1 subunit of NMDAR regulates monocyte transmigration through the brain endothelial cell barrier, J. Neurochem, vol.113, pp.447-453, 2010.

C. D. Sharp, Human Neuroepithelial Cells Express NMDARs, BMC Neurosci, vol.4, p.28, 2003.

G. S. Scott, S. R. Bowman, T. Smith, R. J. Flower, and C. Bolton, Glutamate-stimulated peroxynitrite production in a brain-derived endothelial cell line is dependent on N-methyl-Daspartate (NMDA) receptor activation, Biochem. Pharmacol, vol.73, pp.228-236, 2007.

R. Macrez, Neuroendothelial NMDARs as therapeutic targets in experimental autoimmune encephalomyelitis, Brain, vol.139, pp.2406-2419, 2016.

J. Parcq, Molecular requirements for safer generation of thrombolytics by bioengineering the tissue-type plasminogen activator A chain, J. Thromb. Haemost. JTH, vol.11, pp.539-546, 2013.

R. Macrez, Antibodies Preventing the Interaction of Tissue-Type Plasminogen Activator With N-Methyl-d-Aspartate Receptors Reduce Stroke Damages and Extend the Therapeutic Window of Thrombolysis, Stroke, vol.42, pp.2315-2322, 2011.

O. A. Marcos-contreras, Hyperfibrinolysis increases blood-brain barrier permeability by a plasmin-and bradykinin-dependent mechanism, Blood, vol.128, pp.2423-2434, 2016.

D. S. Bredt and S. H. Snyder, Isolation of nitric oxide synthetase, a calmodulin-requiring enzyme, Proc. Natl. Acad. Sci, vol.87, pp.682-685, 1990.

K. S. Christopherson, B. J. Hillier, W. A. Lim, and D. S. Bredt, PSD-95 Assembles a Ternary Complex with theN-Methyl-d-aspartic Acid Receptor and a Bivalent Neuronal NO Synthase PDZ Domain, J. Biol. Chem, vol.274, pp.27467-27473, 1999.

D. W. Busija, F. Bari, F. Domoki, and T. Louis, Mechanisms Involved in the Cerebrovascular Dilator Effects of N-methyl-D-aspartate in Cerebral Cortex, Brain Res. Rev, vol.56, pp.89-100, 2007.

D. S. Bredt, C. D. Ferris, and S. H. Snyder, Nitric oxide synthase regulatory sites. Phosphorylation by cyclic AMP-dependent protein kinase, protein kinase C, and calcium/calmodulin protein kinase; identification of flavin and calmodulin binding sites, J. Biol. Chem, vol.267, pp.10976-10981, 1992.

Y. Hayashi, Regulation of Neuronal Nitric-oxide Synthase by Calmodulin Kinases, J. Biol. Chem, vol.274, pp.20597-20602, 1999.

G. A. Rameau, L. Chiu, and E. B. Ziff, NMDAR regulation of nNOS phosphorylation and induction of neuron death, Neurobiol. Aging, vol.24, pp.1123-1133, 2003.

G. A. Rameau, Biphasic Coupling of Neuronal Nitric Oxide Synthase Phosphorylation to the NMDAR Regulates AMPA Receptor Trafficking and Neuronal Cell Death, J. Neurosci, vol.27, pp.3445-3455, 2007.

A. Gualandris, T. E. Jones, S. Strickland, and S. E. Tsirka, Membrane depolarization induces calcium-dependent secretion of tissue plasminogen activator, J. Neurosci, vol.16, pp.2220-2225, 1996.

P. T. Pang and B. Lu, Regulation of late-phase LTP and long-term memory in normal and aging hippocampus: role of secreted proteins tPA and BDNF, Ageing Res. Rev, vol.3, pp.407-430, 2004.

D. Baranes, Tissue Plasminogen Activator Contributes to the Late Phase of LTP and to Synaptic Growth in the Hippocampal Mossy Fiber Pathway, Neuron, vol.21, pp.813-825, 1998.

U. Schaefer, T. Machida, S. Vorlova, S. Strickland, and R. Levi, The plasminogen activator system modulates sympathetic nerve function, J. Exp. Med, vol.203, pp.2191-2200, 2006.

S. Lemarchant, tPA promotes ADAMTS-4-induced CSPG degradation, thereby enhancing neuroplasticity following spinal cord injury, Neurobiol. Dis, vol.66, pp.28-42, 2014.

S. S. Segal, Integration and Modulation of Intercellular Signaling Underlying Blood Flow Control, J. Vasc. Res, vol.52, pp.136-157, 2015.

A. Mehra, C. Ali, J. Parcq, D. Vivien, and F. Docagne, The plasminogen activation system in neuroinflammation, Biochim. Biophys. Acta BBA -Mol. Basis Dis, vol.1862, pp.395-402, 2016.

E. Fiumana, H. Parfenova, J. H. Jaggar, and C. W. Leffler, Carbon monoxide mediates vasodilator effects of glutamate in isolated pressurized cerebral arterioles of newborn pigs

, Am. J. Physiol. -Heart Circ. Physiol, vol.284, pp.1073-1079, 2003.

H. Parfenova, A. Fedinec, and C. W. Leffler, Ionotropic Glutamate Receptors in Cerebral Microvascular Endothelium are Functionally Linked to Heme Oxygenase, J. Cereb. Blood Flow Metab, vol.23, pp.190-197, 2003.

J. L. Lemaistre, Coactivation of NMDARs by Glutamate and d-Serine Induces Dilation of Isolated Middle Cerebral Arteries, J. Cereb. Blood Flow Metab, vol.32, pp.537-547, 2012.

L. Lu, Astrocytes drive cortical vasodilatory signaling by activating endothelial NMDARs, J. Cereb. Blood Flow Metab, pp.271678-17734100, 2017.

J. L. Stobart, L. Lu, H. D. Anderson, H. Mori, and C. M. Anderson, Astrocyte-induced cortical vasodilation is mediated by D-serine and endothelial nitric oxide synthase, Proc

, Natl. Acad. Sci. U. S. A, vol.110, pp.3149-3154, 2013.

T. Nassar, In vitro and in vivo effects of tPA and PAI-1 on blood vessel tone, Blood, vol.103, pp.897-902, 2004.

T. Nassar, Regulation of Airway Contractility by Plasminogen Activators through NMethyl-D-Aspartate Receptor-1, Am. J. Respir. Cell Mol. Biol, vol.43, pp.703-711, 2010.

S. N. Heyman, The fibrinolytic system attenuates vascular tone: effects of tissue plasminogen activator (tPA) and aminocaproic acid on renal microcirculation, Br. J. Pharmacol, vol.141, pp.971-978, 2004.

M. J. Cipolla, N. Lessov, and W. M. Clark, Postischemic Attenuation of Cerebral Artery Reactivity Is Increased in the Presence of Tissue Plasminogen Activator, Stroke, vol.31, pp.940-945, 2000.

W. M. Armstead, tPA-S481A Prevents Impairment of Cerebrovascular Autoregulation by Endogenous tPA after Traumatic Brain Injury by Upregulating p38 MAPK and Inhibiting ET-1, J. Neurotrauma, vol.30, pp.1898-1907, 2013.

W. M. Armstead, H. Hekierski, S. Yarovoi, A. A. Higazi, . .-r et al., tPA variant tPA-A296-299 prevents impairment of cerebral autoregulation and necrosis of hippocampal neurons after stroke by inhibiting upregulation of ET-1, J. Neurosci. Res, vol.96, pp.128-137, 2018.

C. Errico, Ultrafast ultrasound localization microscopy for deep super-resolution vascular imaging, Nature, vol.527, pp.499-502, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02007890

E. Tiran, Transcranial Functional Ultrasound Imaging in Freely Moving Awake Mice and Anesthetized Young Rats without Contrast Agent, Ultrasound Med. Biol, vol.43, pp.1679-1689, 2017.

E. Macé, Functional ultrasound imaging of the brain, Nat. Methods, vol.8, pp.662-664, 2011.

W. M. Armstead, J. Riley, J. W. Kiessling, D. B. Cines, A. A. Higazi et al., Novel plasminogen activator inhibitor-1-derived peptide protects against impairment of cerebrovasodilation after photothrombosis through inhibition of JNK MAPK, Am. J. Physiol.-Regul. Integr. Comp. Physiol, vol.299, pp.480-485, 2010.

A. M. Martin, The Functional Role of the Second NPXY Motif of the LRP1 ?-Chain in Tissue-type Plasminogen Activator-mediated Activation of N-Methyl-D-aspartate Receptors

, J. Biol. Chem, vol.283, pp.12004-12013, 2008.

E. G. Levin and G. J. Del-zoppo, Localization of tissue plasminogen activator in the endothelium of a limited number of vessels, Am. J. Pathol, vol.144, pp.855-861, 1994.

C. Stefanitsch, A. E. Lawrence, A. Olverling, I. Nilsson, and L. Fredriksson, tPA Deficiency in Mice Leads to Rearrangement in the Cerebrovascular Tree and Cerebroventricular Malformations, Front. Cell. Neurosci, vol.9, p.456, 2015.

M. Fabiani, Neurovascular coupling in normal aging: A combined optical, ERP and fMRI study, NeuroImage, vol.85, pp.592-607, 2014.

I. Stefanova, Age-related changes of blood-oxygen-level-dependent signal dynamics during optokinetic stimulation, Neurobiol. Aging, vol.34, pp.2277-2286, 2013.

P. Toth, Resveratrol treatment rescues neurovascular coupling in aged mice: role of improved cerebromicrovascular endothelial function and downregulation of NADPH oxidase

, Am. J. Physiol. -Heart Circ. Physiol, vol.306, pp.299-308, 2014.

M. Zaletel, M. Strucl, J. Pretnar-oblak, and B. Zvan, Age-related changes in the relationship between visual evoked potentials and visually evoked cerebral blood flow velocity response

, Funct. Neurol, vol.20, pp.115-120, 2005.

H. Girouard and C. Iadecola, Neurovascular coupling in the normal brain and in hypertension, stroke, and Alzheimer disease, J. Appl. Physiol, vol.100, pp.328-335, 2006.

W. R. Wilkerson and D. C. Sane, Aging and thrombosis, Semin. Thromb. Hemost, vol.28, pp.555-568, 2002.

P. Gaussem, P. Grailhe, and E. Anglés-cano, Sodium dodecyl sulfate-induced dissociation of complexes between human tissue plasminogen activator and its specific inhibitor, J. Biol

, Chem, vol.268, pp.12150-12155, 1993.

A. Boulay, B. Saubaméa, X. Declèves, and M. Cohen-salmon, Purification of Mouse Brain Vessels, J. Vis. Exp. JoVE, vol.53208, 2015.

, whisker stimulation in tPA -/-mice before (?) and after (?) IV injection of rtPA (10mg/kg) and saline, HEPES (0.3M) and MK-801 (0.4mg/kg), rtPA (10mg/kg) and MK-801

, Circles represent values for each mouse (Mean ? SEM, Wilcoxon test, n = 9 for the tPA + saline group, n = 10 for the HEPES + MK-801 group, n = 8 for the rtPA + MK-801 group, n = 9 for the HEPES + AP5 group and n = 8 for the rtPA + AP5 group). (c) Mean CBF signal trace from the S1bf during whisker stimulation of tPA -/-mice before (?) and after (?) the injection of rtPA (10mg/kg) and saline, HEPES (0.3M) and AP5 (0.4mg/kg), or rtPA (10mg/kg) and AP5 (0.4mg/kg)

, HEPES (0.3M) and AP5 (0.4mg/kg), or rtPA (10mg/kg) and AP5 (0.4mg/kg

, = 10 for the HEPES + MK-801 group, n = 8 for the rtPA + MK-801 group, n = 9 for the HEPES + AP5 group and n = 8 for the rtPA + AP5 group). (d) Diagram showing the evolution of the hemodynamic response of each mice between the control condition and after the IV injection of rtPA (10mg/kg) and saline, HEPES (0.3M) and MK-801 (0.4mg/kg), rtPA (10mg/kg) and MK-801 (0.4mg/kg), Doppler speckle images (curves in transparency define the SEM, n = 9 for the rtPA + saline group

, Lines are colored according to whether the response is more than 5% higher (?), between +5% and -5% (?), or decreased for more than 5% (?) after the injection

, Expression of NMDARs is restricted to arteries and arterioles, vol.8

, Only vessels over 10µm in diameter express ?-SMA. In b (scale bar = 100µm), immunostainings reveal GluN1 subunit of the NMDAR (red), ?-SMA (green), laminin (magenta) and cells nuclei (blue). (c) Quantification ?-SMA positive vessels (left) and in ?-SMA negative vessels (right) separately. (d) Quantification of the GluN1 staining fluorescence intensity in ?-SMA positive vessels and in ?-SMA negative vessels (Mean ? SEM, ANOVA test, n = 34 for the ?-SMA + group and n = 29 for the ?-SMA -group, from 4 different mice). (e) Epifluorescence images of isolated vessels from C57BL/6 mice brain revealing Glunomab® staining (red), Col IV (green) and cells nuclei (blue), and corresponding schematic representation. Supplementary figure 1: Doppler Laser speckle imaging allows measurement of the CBF directly through the intact skull. (a) Representative images of a mouse skull, intact (bottom right) or thinned (top right) and corresponding laser Doppler speckle images. (b) Mean CBF 2: Physiological parameters. (a) Schematic representation of the set of physiological parameters, Epifluorescence images of isolated vessels from C57BL/6 mice brain revealing in a (scale bar = 50µ m): PDGF-R? (red), ?-SMA (green) and cells nuclei (blue)