C. F. Barker and R. E. Billingham, Immunologically privileged sites, Adv Immunol, vol.25, pp.1-54, 1977.

Y. Shirai, Transplantation of the rat sarcoma in adult heterogeneous animals, Japan Med World, vol.1, pp.14-19, 1921.

A. Louveau, I. Smirnov, and T. J. Keyes, Structural and functional features of central nervous system lymphatic vessels, Nature, vol.523, pp.337-378, 2015.

A. Aspelund, S. Antila, and S. T. Proulx, A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules, J Exp Med, vol.212, pp.991-1000, 2015.

A. Louveau, Cerebral lymphatic drainage: implication in the brain immune privilege, Med Sci (Paris), vol.31, pp.953-959, 2015.

M. Absinta, S. K. Ha, and G. Nair, Human and nonhuman primate meninges harbor lymphatic vessels that can be visualized noninvasively by MRI, Elife, vol.6, p.29738, 2017.

P. H. Kuo, C. Stuehm, and S. Squire, Meningeal lymphatic vessel flow runs countercurrent to venous flow in the superior sagittal sinus of the human brain, Tomography, vol.4, pp.99-104, 2018.

A. Louveau, J. Herz, and M. N. Alme, CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature, Nat Neurosci, vol.21, pp.1380-91, 2018.

K. Alitalo, The lymphatic vasculature in disease, Nat Med, vol.17, pp.1371-80, 2011.

P. Brouillard, L. Boon, and M. Vikkula, Genetics of lymphatic anomalies, J Clin Invest, vol.124, pp.898-904, 2014.

K. Kerchner, A. Fleischer, and G. Yosipovitch, Lower extremity lymphedema update: pathophysiology, diagnosis, and treatment guidelines, J Am Acad Dermatol, vol.59, pp.324-355, 2008.

K. L. Betterman and N. L. Harvey, The lymphatic vasculature: development and role in shaping immunity, Immunol Rev, vol.271, pp.276-92, 2016.

S. Podgrabinska, O. Kamalu, and L. Mayer, Inflamed lymphatic endothelium suppresses dendritic cell maturation and function via mac-1/ICAM-1-dependent mechanism, J Immunol, vol.183, pp.1767-79, 2009.

U. H. Von-andrian and T. R. Mempel, Homing and cellular traffic in lymph nodes, Nat Rev Immunol, vol.3, pp.867-78, 2003.

S. Antila, S. Karaman, and H. Nurmi, Development and plasticity of meningeal lymphatic vessels, J Exp Med, vol.214, pp.3645-67, 2017.

C. J. Harling-berg, T. J. Park, and P. M. Knopf, Role of the cervical lymphatics in the Th2-type hierarchy of CNS immune regulation, J Neuroimmunol, vol.101, pp.111-138, 1999.

R. O. Carare, C. A. Hawkes, and M. Jeffrey, Review: cerebral amyloid angiopathy, prion angiopathy, CADASIL and the spectrum of protein elimination failure angiopathies (PEFA) in neurodegenerative disease with a focus on therapy, Neuropathol Appl Neurobiol, vol.39, pp.593-611, 2013.

S. Amor, F. Puentes, and D. Baker, Inflammation in neurodegenerative diseases, Immunology, vol.129, pp.154-69, 2010.

T. Brinker, E. Stopa, and J. Morrison, A new look at cerebrospinal fluid circulation, Fluids Barriers CNS, vol.11, p.10, 2014.

N. J. Abbott, Evidence for bulk flow of brain interstitial fluid: significance for physiology and pathology, Neurochem Int, vol.45, pp.545-52, 2004.

N. J. Abbott, M. E. Pizzo, and J. E. Preston, The role of brain barriers in fluid movement in the CNS: is there a 'glymphatic' system?, Acta Neuropathol, vol.135, pp.387-407, 2018.

S. B. Hladky and M. A. Barrand, Mechanisms of fluid movement into, through and out of the brain: evaluation of the evidence, Fluids Barriers CNS, vol.11, p.26, 2014.

Y. Lei, H. Han, and F. Yuan, The brain interstitial system: anatomy, modeling, in vivo measurement, and applications, Prog Neurobiol, vol.157, pp.230-276, 2017.

J. J. Iliff, M. Wang, and Y. Liao, A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta, Sci Transl Med, vol.4, pp.147-111, 2012.

R. O. Carare, M. Bernardes-silva, and T. A. Newman, Solutes, but not cells, drain from the brain parenchyma along basement membranes of capillaries and arteries: significance for cerebral amyloid angiopathy and neuroimmunology, Neuropathol Appl Neurobiol, vol.34, pp.131-175, 2008.

R. R. Lonser, M. Sarntinoranont, and P. F. Morrison, Convectionenhanced delivery to the central nervous system, J Neurosurg, vol.122, pp.697-706, 2015.

I. Szentistvanyi, C. S. Patlak, and R. A. Ellis, Drainage of interstitial fluid from different regions of rat brain, Am J Physiol, vol.246, pp.835-879, 1984.

S. Kida, A. Pantazis, and R. O. Weller, CSF drains directly from the subarachnoid space into nasal lymphatics in the rat. anatomy, histology and immunological significance, Neuropathol Appl Neurobiol, vol.19, pp.480-488, 1993.

S. B. Hladky and M. A. Barrand, Elimination of substances from the brain parenchyma: efflux via perivascular pathways and via the blood-brain barrier, Fluids Barriers CNS, vol.15, p.30, 2018.

P. Giannoni, J. Badaut, and C. Dargazanli, The pericyte-glia interface at the blood-brain barrier, Clin Sci (Lond), vol.132, pp.361-74, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02074159

R. Daneman and A. Prat, The blood-brain barrier, Cold Spring Harb Perspect Biol, vol.7, p.20412, 2015.

N. Marchi, T. Granata, and C. Ghosh, Blood-brain barrier dysfunction and epilepsy: pathophysiologic role and therapeutic approaches, Epilepsia, vol.53, pp.1877-86, 2012.

G. Bar-klein, S. Lublinsky, and L. Kamintsky, Imaging bloodbrain barrier dysfunction as a biomarker for epileptogenesis, Brain, vol.140, pp.1692-705, 2017.

N. J. Abbott and A. Friedman, Overview and introduction: the blood-brain barrier in health and disease, Epilepsia, vol.53, issue.6, pp.1-6, 2012.

F. Frigerio, A. Frasca, and I. Weissberg, Long-lasting pro-ictogenic effects induced in vivo by rat brain exposure to serum albumin in the absence of concomitant pathology, Epilepsia, vol.53, pp.1887-97, 2012.

O. Tomkins, I. Shelef, and I. Kaizerman, Blood-brain barrier disruption in post-traumatic epilepsy, J Neurol Neurosurg Psychiatry, vol.79, pp.774-781, 2008.

N. Maggio, Z. Itsekson, and D. Dominissini, Thrombin regulation of synaptic plasticity: implications for physiology and pathology, Exp Neurol, vol.247, pp.595-604, 2013.

N. Marchi, E. Oby, and A. Batra, In vivo and in vitro effects of pilocarpine: relevance to ictogenesis, Epilepsia, vol.48, pp.1934-1980, 2007.

U. Vazana, R. Veksler, and G. S. Pell, Glutamate-mediated bloodbrain barrier opening: implications for neuroprotection and drug delivery, J Neurosci, vol.36, pp.7727-7766, 2016.

W. Klement, R. Garbelli, and E. Zub, Seizure progression and inflammatory mediators promote pericytosis and pericyte-microglia clustering at the cerebrovasculature, Neurobiol Dis, vol.113, pp.70-81, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02071090

A. Vezzani, B. Lang, and E. Aronica, Immunity and inflammation in epilepsy, Cold Spring Harb Perspect Med, vol.6, p.22699, 2015.

N. Marchi, T. Granata, and D. Janigro, Inflammatory pathways of seizure disorders, Trends Neurosci, vol.37, pp.55-65, 2014.

A. Mendes and L. Sampaio, Brain magnetic resonance in status epilepticus: a focused review, Seizure, vol.38, pp.63-70, 2016.

O. Tomkins, A. Feintuch, and M. Benifla, Blood-brain barrier breakdown following traumatic brain injury: a possible role in posttraumatic epilepsy, Cardiovasc Psychiatry Neurol, p.765923, 2011.

N. Marchi, G. Betto, and V. Fazio, Blood-brain barrier damage and brain penetration of antiepileptic drugs: role of serum proteins and brain edema, Epilepsia, vol.50, pp.664-77, 2009.

B. I. Omalu, S. T. Dekosky, and R. L. Hamilton, Chronic traumatic encephalopathy in a national football league player: part II, Neurosurgery, vol.59, pp.1086-92, 2006.

B. I. Omalu, S. T. Dekosky, and R. L. Minster, Chronic traumatic encephalopathy in a national football league player, Neurosurgery, vol.57, pp.128-162, 2005.

N. Marchi, M. Banjara, and D. Janigro, Blood-brain barrier, bulk flow, and interstitial clearance in epilepsy, J Neurosci Methods, vol.260, pp.118-142, 2016.

T. Makinen, L. Jussila, and T. Veikkola, Inhibition of lymphangiogenesis with resulting lymphedema in transgenic mice expressing soluble VEGF receptor-3, Nat Med, vol.7, pp.199-205, 2001.

B. A. Plog, K. M. Moll, and H. Kang, A novel technique for morphometric quantification of subarachnoid hemorrhage-induced microglia activation, J Neurosci Methods, vol.229, pp.44-52, 2014.

I. Ali, J. C. Silva, and S. Liu, Targeting neurodegeneration to prevent post-traumatic epilepsy, Neurobiol Dis, vol.123, pp.100-109, 2019.

P. G. Saletti, I. Ali, and P. M. Casillas-espinosa, In search of antiepileptogenic treatments for post-traumatic epilepsy, Neurobiol Dis, vol.123, pp.86-99, 2019.

P. Zheng, S. R. Shultz, and C. M. Hovens, Hyperphosphorylated tau is implicated in acquired epilepsy and neuropsychiatric comorbidities, Mol Neurobiol, vol.49, pp.1532-1541, 2014.

H. B. Sarnat and L. Flores-sarnat, Infantile tauopathies: hemimegalencephaly; tuberous sclerosis complex; focal cortical dysplasia 2; ganglioglioma, Brain Dev, vol.37, pp.553-62, 2015.

A. Sen, M. Thom, and L. Martinian, Pathological tau tangles localize to focal cortical dysplasia in older patients, Epilepsia, vol.48, pp.1447-54, 2007.

M. Thom, J. Y. Liu, and P. Thompson, Neurofibrillary tangle pathology and braak staging in chronic epilepsy in relation to traumatic brain injury and hippocampal sclerosis: a post-mortem study, Brain, vol.134, pp.2969-81, 2011.

V. Puvenna, M. Engeler, and M. Banjara, Is phosphorylated tau unique to chronic traumatic encephalopathy? phosphorylated tau in epileptic brain and chronic traumatic encephalopathy, Brain Res, vol.1630, pp.225-265, 2016.

G. Monti, M. Tondelli, and G. Giovannini, Cerebrospinal fluid tau proteins in status epilepticus, Epilepsy Behav, vol.49, pp.150-154, 2015.

Z. Liang, F. Liu, and K. Iqbal, Dysregulation of tau phosphorylation in mouse brain during excitotoxic damage, J Alzheimers Dis, vol.17, pp.531-540, 2009.

B. E. Hawkins, S. Krishnamurthy, and D. L. Castillo-carranza, Rapid accumulation of endogenous tau oligomers in a rat model of traumatic brain injury: possible link between traumatic brain injury and sporadic tauopathies, J Biol Chem, vol.288, pp.17042-50, 2013.

S. R. Shultz, D. K. Wright, and P. Zheng, Sodium selenate reduces hyperphosphorylated tau and improves outcomes after traumatic brain injury, Brain, vol.138, pp.1297-313, 2015.

E. D. Roberson, B. Halabisky, and J. W. Yoo, Amyloid-beta/fyninduced synaptic, network, and cognitive impairments depend on tau levels in multiple mouse models of alzheimer's disease, J Neurosci, vol.31, pp.700-711, 2011.

J. M. Decker, L. Kruger, and A. Sydow, The tau/A152T mutation, a risk factor for frontotemporal-spectrum disorders, leads to NR2B receptor-mediated excitotoxicity, EMBO Rep, vol.17, pp.552-69, 2016.

L. Kandratavicius, M. R. Monteiro, and J. E. Hallak, Microtubuleassociated proteins in mesial temporal lobe epilepsy with and without psychiatric comorbidities and their relation with granular cell layer dispersion, Biomed Res Int, p.960126, 2013.

I. Sotiropoulos, M. C. Galas, and J. M. Silva, Atypical, non-standard functions of the microtubule associated tau protein, Acta Neuropathol Commun, vol.5, p.91, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01833336

T. Engel, P. Goni-oliver, and J. J. Lucas, Chronic lithium administration to FTDP-17 tau and GSK-3beta overexpressing mice prevents tau hyperphosphorylation and neurofibrillary tangle formation, but pre-formed neurofibrillary tangles do not revert, J Neurochem, vol.99, pp.1445-55, 2006.

M. Bhowmik, R. Khanam, and N. Saini, Activation of AKT/ GSK3beta pathway by TDZD-8 attenuates kainic acid induced neurodegeneration but not seizures in mice, Neurotoxicology, vol.46, pp.44-52, 2015.

N. C. Jones, T. Nguyen, and N. M. Corcoran, Targeting hyperphosphorylated tau with sodium selenate suppresses seizures in rodent models, Neurobiol Dis, vol.45, pp.897-901, 2012.

S. J. Liu, P. Zheng, and D. K. Wright, Sodium selenate retards epileptogenesis in acquired epilepsy models reversing changes in protein phosphatase 2A and hyperphosphorylated tau, Brain, vol.139, pp.1919-1957, 2016.

L. Wilson, W. Stewart, -. Dams, and K. Connor, The chronic and evolving neurological consequences of traumatic brain injury, Lancet Neurol, vol.16, pp.813-838, 2017.

M. Molteni and C. Rossetti, Neurodegenerative diseases: the immunological perspective, J Neuroimmunol, vol.313, pp.109-124, 2017.

H. E. Gendelman and R. L. Mosley, A perspective on roles played by innate and adaptive immunity in the pathobiology of neurodegenerative disorders, J Neuroimmune Pharmacol, vol.10, pp.645-50, 2015.

J. Bauer, A. J. Becker, and W. Elyaman, Innate and adaptive immunity in human epilepsies, Epilepsia, vol.58, issue.3, pp.57-68, 2017.

J. T. Walsh, N. Watson, and J. Kipnis, T cells in the central nervous system: messengers of destruction or purveyors of protection?, Immunology, vol.141, pp.340-344, 2014.

X. Urra, F. Miro, and A. Chamorro, Antigen-specific immune reactions to ischemic stroke, Front Cell Neurosci, vol.8, p.278, 2014.

A. F. De-vos, M. Van-meurs, and H. P. Brok, Transfer of central nervous system autoantigens and presentation in secondary lymphoid organs, J Immunol, vol.169, pp.5415-5438, 2002.

V. H. Perry, A revised view of the central nervous system microenvironment and major histocompatibility complex class II antigen presentation, J Neuroimmunol, vol.90, pp.113-134, 1998.

R. O. Weller, I. Galea, and R. O. Carare, Pathophysiology of the lymphatic drainage of the central nervous system: implications for pathogenesis and therapy of multiple sclerosis, Pathophysiology, vol.17, pp.295-306, 2010.

B. Engelhardt, R. O. Carare, and I. Bechmann, Vascular, glial, and lymphatic immune gateways of the central nervous system, Acta Neuropathol, vol.132, pp.317-355, 2016.

H. F. Cserr, C. J. Harling-berg, and P. M. Knopf, Drainage of brain extracellular fluid into blood and deep cervical lymph and its immunological significance, Brain Pathol, vol.2, pp.269-76, 1992.

D. L. Thomas, D. M. Kranz, and E. J. Roy, Experimental manipulations of afferent immune responses influence efferent immune responses to brain tumors, Cancer Immunol Immunother, vol.57, pp.1323-1356, 2008.

J. T. Walsh, J. Zheng, and I. Smirnov, Regulatory T cells in central nervous system injury: a double-edged sword, J Immunol, vol.193, pp.5013-5035, 2014.

H. Widner, G. Moller, and B. B. Johansson, Immune response in deep cervical lymph nodes and spleen in the mouse after antigen deposition in different intracerebral sites, Scand J Immunol, vol.28, pp.563-71, 1988.

I. Galea, I. Bechmann, and V. H. Perry, What is immune privilege (not)?, Trends Immunol, vol.28, pp.12-20, 2007.

I. E. Scheffer, S. Berkovic, and G. Capovilla, ILAE classification of the epilepsies: position paper of the ILAE commission for classification and terminology, Epilepsia, vol.58, pp.512-533, 2017.

E. Lancaster, The diagnosis and treatment of autoimmune encephalitis, J Clin Neurol, vol.12, pp.1-13, 2016.

A. Vincent, S. R. Irani, and B. Lang, The growing recognition of immunotherapy-responsive seizure disorders with autoantibodies to specific neuronal proteins, Curr Opin Neurol, vol.23, pp.144-50, 2010.

S. R. Irani, A. Vincent, and J. M. Schott, Autoimmune encephalitis, BMJ, vol.342, 1918.

T. Rasmussen, J. Olszewski, and D. Lloydsmith, Focal seizures due to chronic localized encephalitis, Neurology, vol.8, pp.435-480, 1958.

C. A. Pardo, R. Nabbout, and A. S. Galanopoulou, Mechanisms of epileptogenesis in pediatric epileptic syndromes: rasmussen encephalitis, infantile spasms, and febrile infection-related epilepsy syndrome (FIRES), Neurotherapeutics, vol.11, pp.297-310, 2014.

Y. Robitaille, Chronic encephalitis and epilepsy. Rasmussen's syndrome Ed, pp.79-110, 1991.

S. Varadkar, C. G. Bien, and C. A. Kruse, Rasmussen's encephalitis: clinical features, pathobiology, and treatment advances, Lancet Neurol, vol.13, pp.195-205, 2014.

Y. Li, A. Uccelli, and K. D. Laxer, Local-clonal expansion of infiltrating T lymphocytes in chronic encephalitis of rasmussen, J Immunol, vol.158, pp.1428-1465, 1997.

N. Schwab, C. G. Bien, and A. Waschbisch, CD8 + T-cell clones dominate brain infiltrates in rasmussen encephalitis and persist in the periphery, Brain, vol.132, pp.1236-1282, 2009.

A. Nimer, F. Jelcic, I. Kempf, and C. , Phenotypic and functional complexity of brain-infiltrating T cells in rasmussen encephalitis, Neurol Neuroimmunol Neuroinflamm, vol.5, p.419, 2017.

T. Granata, F. Andermann, and . Rasmussen, Handb Clin Neurol, vol.111, pp.511-520, 2013.

C. G. Bien, J. Bauer, and T. L. Deckwerth, Destruction of neurons by cytotoxic T cells: a new pathogenic mechanism in rasmussen's encephalitis, Ann Neurol, vol.51, pp.311-319, 2002.

J. Bauer, A. Vezzani, and C. G. Bien, Epileptic encephalitis: the role of the innate and adaptive immune system, Brain Pathol, vol.22, pp.412-433, 2012.

S. M. Stamatovic, R. F. Keep, and A. V. Andjelkovic, Brain endothelial cell-cell junctions: how to "open" the blood brain barrier, Curr Neuropharmacol, vol.6, pp.179-92, 2008.

G. Haraldsen, D. Kvale, and B. Lien, Cytokine-regulated expression of E-selectin, intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) in human microvascular endothelial cells, J Immunol, vol.156, pp.2558-65, 1996.

H. Kim, R. P. Kataru, and G. Y. Koh, Inflammation-associated lymphangiogenesis: a double-edged sword?, J Clin Invest, vol.124, pp.936-978, 2014.

R. P. Kataru, K. Jung, and C. Jang, Critical role of CD11b+ macrophages and VEGF in inflammatory lymphangiogenesis, antigen clearance, and inflammation resolution, Blood, vol.113, pp.5650-5659, 2009.

R. Huggenberger, S. S. Siddiqui, and D. Brander, An important role of lymphatic vessel activation in limiting acute inflammation, Blood, vol.117, pp.4667-78, 2011.

R. Guo, Q. Zhou, and S. T. Proulx, Inhibition of lymphangiogenesis and lymphatic drainage via vascular endothelial growth factor receptor 3 blockade increases the severity of inflammation in a mouse model of chronic inflammatory arthritis, Arthritis Rheum, vol.60, pp.2666-76, 2009.

C. T. Kesler, S. Liao, and L. L. Munn, Lymphatic vessels in health and disease, Wiley Interdiscip Rev Syst Biol Med, vol.5, pp.111-135, 2013.

A. Szuba, M. Skobe, and M. J. Karkkainen, Therapeutic lymphangiogenesis with human recombinant VEGF-C, FASEB J, vol.16, pp.1985-1992, 2002.

T. Tammela, A. Saaristo, and T. Holopainen, Therapeutic differentiation and maturation of lymphatic vessels after lymph node dissection and transplantation, Nat Med, vol.13, pp.1458-66, 2007.

M. T. Visuri, K. M. Honkonen, and P. Hartiala, VEGF-C and VEGF-C156S in the pro-lymphangiogenic growth factor therapy of lymphedema: a large animal study, Angiogenesis, vol.18, pp.313-339, 2015.

S. Karaman, H. Nurmi, and S. Antila, Stimulation and inhibition of lymphangiogenesis via adeno-associated viral gene delivery, Methods Mol Biol, vol.1846, pp.291-300, 2018.