N. J. Abbott, A. A. Patabendige, D. E. Dolman, S. R. Yusof, and D. J. Begley, Structure and function of the blood-brain barrier, Neurobiol. Dis, vol.37, pp.13-25, 2010.

J. B. Adams, L. J. Johansen, L. D. Powell, D. Quig, and R. A. Rubin, Gastrointestinal flora and gastrointestinal status in children with autismcomparisons to typical children and correlation with autism severity, BMC Gastroenterol, vol.11, p.22, 2011.

A. Aggarwal, A. Khera, I. Singh, and R. Sandhir, S-nitrosoglutathione prevents blood-brain barrier disruption associated with increased matrix metalloproteinase-9 activity in experimental diabetes, J. Neurochem, vol.132, pp.595-608, 2015.

G. Aimaretti, M. R. Ambrosio, C. Di-somma, M. Gasperi, S. Cannavò et al., Residual pituitary function after brain injury-induced hypopituitarism: a prospective 12-month study, J. Clin. Endocrinol. Metab, vol.90, pp.6085-6092, 2005.

H. B. Allen, Alzheimer's disease: assessing the role of spirochetes, biofilms, the immune system, and amyloid-? with regard to potential treatment and prevention, J. Alzheimers Dis, vol.27, pp.1271-1276, 2016.

R. Alonso, D. Pisa, A. I. Marina, E. Morato, A. Rábano et al., Fungal infection in patients with Alzheimer's disease, J. Alzheimers Dis, vol.41, pp.301-311, 2014.

A. Amedei and F. Boem, I've gut a feeling: microbiota impacting the conceptual and experimental perspectives of personalized medicine, Int. J. Mol. Sci, vol.19, p.3756, 2018.

M. Arango-lievano, B. Boussadia, L. D. De-terdonck, C. Gault, P. Fontanaud et al., Topographic reorganization of cerebrovascular mural cells under seizure conditions, Cell Rep, vol.24, pp.1045-1059, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02071076

A. Armulik, G. Genové, M. Mäe, M. H. Nisancioglu, E. Wallgard et al., Pericytes regulate the blood-brain barrier, Nature, vol.468, pp.557-561, 2017.

A. Aspelund, S. Antila, S. T. Proulx, T. V. Karlsen, S. Karaman et al., A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules, J. Exp. Med, vol.212, pp.991-999, 2015.

A. A. Asuni, A. Boutajangout, D. Quartermain, and E. M. Sigurdsson, Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements, J. Neurosci, vol.27, pp.9115-9129, 2007.

J. Bach and R. Dodel, Naturally occurring autoantibodies against ?-Amyloid, Adv. Exp. Med. Biol, vol.750, pp.91-99, 2012.

B. J. Balin, H. C. Gérard, E. J. Arking, D. M. Appelt, P. J. Branigan et al., Identification and localization of Chlamydia pneumoniae in the Alzheimer's brain, Med. Microbiol. Immunol, vol.187, pp.23-42, 1998.

A. Bartos, L. Fialová, J. Svarcová, and D. Ripova, Patients with Alzheimer disease have elevated intrathecal synthesis of antibodies against tau protein and heavy neurofilament, J. Neuroimmunol, vol.252, pp.100-105, 2012.

R. D. Bell, E. A. Winkler, A. P. Sagare, I. Singh, B. Larue et al., Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging, Neuron, vol.68, pp.409-427, 2010.

K. Blennow, J. Hardy, and H. Zetterberg, The neuropathology and neurobiology of traumatic brain injury, Neuron, vol.76, pp.886-899, 2012.

V. Braniste, M. Al-asmakh, C. Kowal, F. Anuar, A. Abbaspour et al., The gut microbiota influences blood-brain barrier permeability in mice, Sci. Transl. Med, vol.6, pp.263-158, 2014.

S. Brettschneider, N. G. Morgenthaler, S. J. Teipel, C. Fischer-schulz, K. Bürger et al., Decreased serum amyloid beta(1-42) autoantibody levels in Alzheimer's disease, determined by a newly developed immunoprecipitation assay with radiolabeled amyloid beta(1-42) peptide, Biol. Psychiatry, vol.57, pp.813-816, 2005.

M. Britschgi, C. E. Olin, H. T. Johns, Y. Takeda-uchimura, M. C. Lemieux et al., Neuroprotective natural antibodies to assemblies of amyloidogenic peptides decrease with normal aging and advancing Alzheimer's disease, Proc. Natl. Acad. Sci. U.S.A, vol.106, pp.12145-12150, 2009.

S. Busse, M. Busse, B. Brix, C. Probst, A. Genz et al., Seroprevalence of N-methyl-D-aspartate glutamate receptor (NMDA-R) autoantibodies in aging subjects without neuropsychiatric disorders and in dementia patients, Eur. Arch. Psychiatry Clin. Neurosci, vol.264, pp.545-550, 2014.

M. I. Butler, J. F. Cryan, and T. G. Dinan, Man and the microbiome: a new theory of everything?, Annu. Rev. Clin. Psychol, vol.15, pp.371-398, 2019.

P. Casali and E. W. Schettino, Structure and function of natural antibodies, Curr. Top. Microbiol. Immunol, vol.210, pp.167-179, 1996.

A. Cattaneo, N. Cattane, S. Galluzzi, S. Provasi, N. Lopizzo et al., Association of brain amyloidosis with pro-inflammatory gut bacterial taxa and peripheral inflammation markers in cognitively impaired elderly, Neurobiol. Aging, vol.49, pp.60-68, 2017.

M. Cerovic, G. Forloni, and C. Balducci, Neuroinflammation and the gut microbiota: possible alternative therapeutic targets to counteract Alzheimer's disease? Front, Aging Neurosci, vol.11, p.284, 2019.

C. Chu, M. H. Murdock, D. Jing, T. H. Won, H. Chung et al., The microbiota regulate neuronal function and fear extinction learning, Nature, vol.574, pp.543-548, 2019.

L. M. Cox and H. L. Weiner, Microbiota signaling pathways that influence neurologic disease, Neurother. J. Am. Soc. Exp. Neurother, vol.15, pp.135-145, 2018.

E. Cristante, S. Mcarthur, C. Mauro, E. Maggioli, I. A. Romero et al., Identification of an essential endogenous regulator of blood-brain barrier integrity, and its pathological and therapeutic implications, Proc. Natl. Acad. Sci. U.S.A, vol.110, pp.832-841, 2013.

J. F. Cryan, K. J. O'riordan, C. S. Cowan, K. V. Sandhu, T. F. Bastiaanssen et al., The microbiota-gut-brain axis, Physiol. Rev, vol.99, pp.1877-2013, 2019.

R. Daneman, L. Zhou, A. A. Kebede, and B. A. Barres, Pericytes are required for blood-brain barrier integrity during embryogenesis, Nature, vol.468, pp.562-566, 2010.

T. V. Davydova, N. I. Voskresenskaya, V. G. Fomina, L. A. Vetrile, and O. A. Doronina, Induction of autoantibodies to glutamate in patients with Alzheimer's disease, Bull. Exp. Biol. Med, vol.143, pp.182-183, 2007.

R. Deane, S. Du-yan, R. K. Submamaryan, B. Larue, S. Jovanovic et al., RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain, Nat. Med, vol.9, pp.907-913, 2003.

R. Deane, I. Singh, A. P. Sagare, R. D. Bell, N. T. Ross et al., A multimodal RAGE-specific inhibitor reduces amyloid ?-mediated brain disorder in a mouse model of Alzheimer disease, J. Clin. Invest, vol.122, pp.1377-1392, 2012.

M. B. Dinkins, S. Dasgupta, G. Wang, G. Zhu, Q. He et al., The 5XFAD mouse model of Alzheimer's disease exhibits an age-dependent increase in anti-ceramide IgG and exogenous administration of ceramide further increases anti-ceramide titers and amyloid plaque burden, J. Alzheimers Dis, vol.46, pp.55-61, 2015.

R. Dodel, K. Balakrishnan, K. Keyvani, O. Deuster, F. Neff et al., Naturally occurring autoantibodies against beta-amyloid: investigating their role in transgenic animal and in vitro models of Alzheimer's disease, J. Neurosci, vol.31, pp.5847-5854, 2011.

S. S. Dominy, C. Lynch, F. Ermini, M. Benedyk, A. Marczyk et al., Porphyromonas gingivalis in Alzheimer's disease brains: evidence for disease causation and treatment with small-molecule inhibitors, Sci. Adv, vol.5, p.3333, 2019.

Y. Du, R. Dodel, H. Hampel, K. Buerger, S. Lin et al., Reduced levels of amyloid beta-peptide antibody in Alzheimer disease, Neurology, vol.57, pp.801-805, 2001.

W. A. Eimer, D. K. Vijaya-kumar, N. K. Navalpur-shanmugam, A. S. Rodriguez, T. Mitchell et al., Alzheimer's disease-associated ?amyloid is rapidly seeded by herpesviridae to protect against brain infection, Neuron, vol.11, pp.3-63, 2018.

K. B. Elkon and G. J. Silverman, Naturally occurring autoantibodies to apoptotic cells, Adv. Exp. Med. Biol, vol.750, pp.14-26, 2012.

B. Engelhardt, P. Vajkoczy, and R. O. Weller, The movers and shapers in immune privilege of the CNS, Nat. Immunol, vol.18, pp.123-131, 2017.

L. Fialová, A. Bartos, J. Svarcová, and I. Malbohan, Increased intrathecal high-avidity anti-tau antibodies in patients with multiple sclerosis, PLoS One, vol.6, p.27476, 2011.

T. C. Fung, C. A. Olson, and E. Y. Hsiao, Interactions between the microbiota, immune and nervous systems in health and disease, Nat. Neurosci, vol.20, pp.145-155, 2017.

P. Giannoni, J. Badaut, C. Dargazanli, A. F. De-maudave, W. Klement et al., The pericyte-glia interface at the blood-brain barrier, Clin. Sci. Lond. Engl, vol.14, pp.361-374, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02074159

L. E. Goldstein, A. M. Fisher, C. A. Tagge, X. Zhang, L. Velisek et al., Tetramethylpyrazine attenuates blood-brain barrier disruption in ischemia/reperfusion injury through the JAK/STAT signaling pathway, :134ra60. Gong, vol.4, pp.289-297, 2012.

A. V. Goryunova, N. A. Bazarnaya, E. G. Sorokina, N. Y. Semenova, O. V. Globa et al., Glutamate receptor autoantibody concentrations in children with chronic post-traumatic headache, Neurosci. Behav. Physiol, vol.37, pp.761-764, 2007.

M. A. Gruden, T. B. Davidova, M. Malisauskas, R. D. Sewell, N. I. Voskresenskaya et al., Differential neuroimmune markers to the onset of Alzheimer's disease neurodegeneration and dementia: autoantibodies to Abeta((25-35)) oligomers, S100b and neurotransmitters, J. Neuroimmunol, vol.186, pp.181-192, 2007.

K. A. Gustaw, M. R. Garrett, H. Lee, R. J. Castellani, M. G. Zagorski et al., Antigen-antibody dissociation in Alzheimer disease: a novel approach to diagnosis, J. Neurochem, vol.106, pp.1350-1356, 2008.

T. Harach, N. Marungruang, N. Duthilleul, V. Cheatham, K. D. Mc-coy et al., Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota, Sci. Rep, vol.08, p.41802, 2017.

W. H. Hind, T. J. England, and S. E. Sullivan, Cannabidiol protects an in vitro model of the blood-brain barrier from oxygen-glucose deprivation via PPAR? and 5-HT1A receptors, Br. J. Pharmacol, vol.173, pp.815-825, 2016.

S. Hong, H. Jiancheng, W. Jiawen, Z. Shuqin, Z. Guilian et al., Losartan inhibits development of spontaneous recurrent seizures by preventing astrocyte activation and attenuating blood-brain barrier permeability following pilocarpine-induced status epilepticus, Brain Res. Bull, vol.149, pp.251-259, 2019.

C. D. Hue, F. S. Cho, S. Cao, . Dale, C. R. Bass et al., Dexamethasone potentiates in vitro blood-brain barrier recovery after primary blast injury by glucocorticoid receptor-mediated upregulation of ZO-1 tight junction protein, J. Cereb. Blood Flow Metab, vol.35, pp.1191-1198, 2015.

C. Iadecola, F. Zhang, K. Niwa, C. Eckman, S. K. Turner et al., SOD1 rescues cerebral endothelial dysfunction in mice overexpressing amyloid precursor protein, Nat. Neurosci, vol.2, pp.157-161, 1999.

R. F. Itzhaki, R. Lathe, B. J. Balin, M. J. Ball, E. L. Bearer et al., Microbes and Alzheimer's disease, J. Alzheimers Dis, vol.51, pp.979-984, 2016.

C. A. Janeway, P. Travers, M. Walport, and M. J. Shlomchik, , 2001.

. Immunobiology and . Edn,

D. Kang, J. B. Adams, D. M. Coleman, E. L. Pollard, J. Maldonado et al., Long-term benefit of microbiota transfer therapy on autism symptoms and gut microbiota, Sci. Rep, vol.9, p.5821, 2019.

P. Karczewski, P. Hempel, R. Kunze, and M. Bimmler, Agonistic autoantibodies to the ?(1) -adrenergic receptor and the ?(2) -adrenergic receptor in Alzheimer's and vascular dementia, Scand. J. Immunol, vol.75, pp.524-530, 2012.

M. Kaya, R. Kalayci, M. Küçük, N. Arican, I. Elmas et al., Effect of losartan on the blood-brain barrier permeability in diabetic hypertensive rats, Life Sci, vol.73, pp.3235-3244, 2003.

A. Kellner, J. Matschke, C. Bernreuther, H. Moch, I. Ferrer et al., Autoantibodies against beta-amyloid are common in Alzheimer's disease and help control plaque burden, Ann. Neurol, vol.65, pp.24-31, 2009.

J. R. Kelly, Y. Borre, C. O'-brien, E. Patterson, S. El-aidy et al., Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat, J. Psychiatr. Res, vol.82, pp.109-118, 2016.

D. L. Kenney-jung, A. Vezzani, R. J. Kahoud, R. G. Lafrance-corey, M. Ho et al., Febrile infection-related epilepsy syndrome treated with anakinra, Ann. Neurol, vol.80, pp.939-945, 2016.

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.

W. Klement, M. Blaquiere, E. Zub, F. Debock, F. Boux et al., A pericyte-glia scarring develops at the leaky capillaries in the hippocampus during seizure activity, Epilepsia, vol.60, pp.1399-1411, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02335992

F. H. Kobeissy, Brain Neurotrauma: Molecular, Neuropsychological, and Rehabilitation Aspects, 2015.

A. Kondo, K. Shahpasand, R. Mannix, J. Qiu, J. Moncaster et al., Antibody against early driver of neurodegeneration cis P-tau blocks brain injury and tauopathy, Nature, vol.523, pp.431-436, 2015.

H. H. Kornhuber, Propionibacterium acnes in the cortex of patients with Alzheimer's disease, Eur. Arch. Psychiatry Clin. Neurosci, vol.246, pp.108-109, 1996.

R. Kosaraju, W. Guesdon, M. J. Crouch, H. L. Teague, E. M. Sullivan et al., B cell activity is impaired in human and mouse obesity and is responsive to an essential fatty acid upon murine influenza infection, J. Immunol, vol.198, pp.4738-4752, 2017.

J. Kountouras, M. Tsolaki, E. Gavalas, M. Boziki, C. Zavos et al., Relationship between Helicobacter pylori infection and Alzheimer disease, Neurology, vol.66, pp.938-940, 2006.

Y. Kronimus, A. Albus, M. Balzer-geldsetzer, S. Straub, E. Semler et al., Naturally occurring autoantibodies against tau protein are reduced in Parkinson's disease dementia, PLoS One, vol.11, 2016.

M. Kucuk, M. Kaya, R. Kalayci, V. Cimen, H. Kudat et al., Effects of losartan on the blood-brain barrier permeability in long-term nitric oxide blockade-induced hypertensive rats, Life Sci, vol.71, pp.1772-1773, 2002.

D. K. Kumar, S. H. Choi, K. J. Washicosky, W. A. Eimer, S. Tucker et al., Amyloid-? peptide protects against microbial infection in mouse and worm models of Alzheimer's disease, Sci. Transl. Med, vol.8, pp.340-72, 2016.

N. Kumar and S. C. Forster, Genome watch: microbiota shuns the modern world, Nat. Rev. Microbiol, vol.15, pp.710-710, 2017.

D. Lazic, A. P. Sagare, A. M. Nikolakopoulou, J. H. Griffin, R. Vassar et al., 3K3A-activated protein C blocks amyloidogenic BACE1 pathway and improves functional outcome in mice, J. Exp. Med, vol.216, pp.279-293, 2019.

L. Page, A. Dupuis, G. Frost, E. H. Larbi, A. Pawelec et al., Role of the peripheral innate immune system in the development of Alzheimer's disease, Exp. Gerontol, vol.01, pp.59-66, 2018.

Y. K. Lee, J. S. Menezes, Y. Umesaki, and S. K. Mazmanian, , 2011.

, Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis, Proc. Natl. Acad. Sci. U.S.A, vol.108, pp.4615-4622

B. Lelouvier, F. Servant, S. Païssé, A. Brunet, S. Benyahya et al., Changes in blood microbiota profiles associated with liver fibrosis in obese patients: a pilot analysis, Hepatology, vol.64, pp.2015-2027, 2016.

E. C. Levin, N. K. Acharya, M. Han, S. B. Zavareh, J. C. Sedeyn et al., Brain-reactive autoantibodies are nearly ubiquitous in human sera and may be linked to pathology in the context of blood-brain barrier breakdown, Brain Res, vol.1345, pp.221-232, 2010.

W. Lin, M. A. Wozniak, R. J. Cooper, G. K. Wilcock, and R. F. Itzhaki, Herpesviruses in brain and Alzheimer's disease, J. Pathol, vol.197, pp.395-402, 2002.

M. Lindhagen-persson, K. Brännström, M. Vestling, M. Steinitz, and A. Olofsson, Amyloid-? oligomer specificity mediated by the IgM isotypeimplications for a specific protective mechanism exerted by endogenous autoantibodies, PLoS One, vol.5, p.13928, 2010.

A. Louveau, T. H. Harris, and J. Kipnis, Revisiting the mechanisms of CNS immune privilege, Trends Immunol, vol.36, pp.569-577, 2015.

A. Louveau, J. Herz, M. N. Alme, A. F. Salvador, M. Q. Dong et al., CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature, Nat. Neurosci, vol.21, pp.1380-1391, 2018.

A. Louveau, I. Smirnov, T. J. Keyes, J. D. Eccles, S. J. Rouhani et al., Structural and functional features of central nervous system lymphatic vessels, Nature, vol.523, pp.337-341, 2015.

C. A. Lozupone, J. I. Stombaugh, J. I. Gordon, J. K. Jansson, and R. Knight, Diversity, stability and resilience of the human gut microbiota, Nature, vol.489, pp.220-230, 2012.

P. Lyden, K. E. Pryor, C. S. Coffey, M. Cudkowicz, R. Conwit et al., Final results of the RHAPSODY trial: a multi-center, phase 2 trial using a continual reassessment method to determine the safety and tolerability of 3K3A-APC, a recombinant variant of human activated protein C, in combination with tissue plasminogen activator, mechanical thrombectomy or both in moderate to severe acute ischemic stroke, Ann. Neurol, vol.85, pp.125-136, 2019.

M. Maftei, F. Thurm, C. Schnack, H. Tumani, M. Otto et al., Increased levels of antigen-bound ?-amyloid autoantibodies in serum and cerebrospinal fluid of Alzheimer's disease patients, PLoS One, vol.8, p.68996, 2013.

R. Mannix, W. P. Meehan, J. Mandeville, P. E. Grant, T. Gray et al., Clinical correlates in an experimental model of repetitive mild brain injury, Ann. Neurol, vol.74, pp.65-75, 2013.

N. Marchi, L. Angelov, T. Masaryk, V. Fazio, T. Granata et al., Seizure-promoting effect of blood-brain barrier disruption, Epilepsia, vol.48, pp.732-742, 2007.

N. Marchi, J. J. Bazarian, V. Puvenna, M. Janigro, C. Ghosh et al., Consequences of repeated blood-brain barrier disruption in football players, PLoS One, vol.8, p.56805, 2013.

N. Marchi, Q. Fan, C. Ghosh, V. Fazio, F. Bertolini et al., Antagonism of peripheral inflammation reduces the severity of status epilepticus, Neurobiol. Dis, vol.33, pp.171-181, 2009.

N. Marchi, T. Granata, E. Freri, E. Ciusani, F. Ragona et al., Efficacy of anti-inflammatory therapy in a model of acute seizures and in a population of pediatric drug resistant epileptics, PLoS One, vol.6, p.18200, 2011.

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

C. R. Martin, V. Osadchiy, A. Kalani, and E. A. Mayer, The brain-gutmicrobiome axis, Cell Mol. Gastroenterol. Hepatol, vol.6, pp.133-148, 2018.

A. Marushima, M. Nieminen, I. Kremenetskaia, R. Gianni-barrera, J. Woitzik et al., Balanced single-vector co-delivery of VEGF/PDGF-BB improves functional collateralization in chronic cerebral ischemia, J. Cereb. Blood Flow Metab, vol.9, pp.271678-18818298, 2019.

J. A. Mcintyre, J. Chapman, E. Shavit, R. L. Hamilton, and S. T. Dekosky, Redox-reactive autoantibodies in Alzheimer's patients' cerebrospinal fluids: preliminary studies, Autoimmunity, vol.40, pp.390-396, 2007.

J. A. Mcintyre, C. J. Ramsey, B. D. Gitter, A. J. Saykin, D. R. Wagenknecht et al., Antiphospholipid autoantibodies as blood biomarkers for detection of early stage Alzheimer's disease, Autoimmunity, vol.48, pp.344-351, 2015.

A. Mcrae, R. N. Martins, J. Fonte, R. Kraftsik, L. Hirt et al., Cerebrospinal fluid antimicroglial antibodies in Alzheimer disease: a putative marker of an ongoing inflammatory process, Exp. Gerontol, vol.42, pp.355-363, 2007.

P. Mecocci, L. Parnetti, G. Romano, A. Scarelli, F. Chionne et al., Serum anti-GFAP and anti-S100 autoantibodies in brain aging, Alzheimer's disease and vascular dementia, J. Neuroimmunol, vol.57, pp.165-170, 1995.

J. Miklossy, Analysis of the evidence following Koch's and Hill's criteria, J. Neuroinflamm, vol.8, p.90, 2011.

J. Miklossy, Bacterial amyloid and DNA are important constituents of senile plaques: further evidence of the spirochetal and biofilm nature of senile plaques, J. Alzheimers Dis, vol.13, pp.1459-1473, 2016.

M. R. Minter, R. Hinterleitner, M. Meisel, C. Zhang, V. Leone et al., Antibiotic-induced perturbations in microbial diversity during post-natal development alters amyloid pathology in an aged APPSWE/PS1 E9 murine model of Alzheimer's disease, Sci. Rep, vol.7, p.10411, 2017.

M. R. Minter, C. Zhang, V. Leone, D. L. Ringus, X. Zhang et al., Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of Alzheimer's disease, Sci. Rep, vol.21, p.30028, 2016.

R. D. Moir, R. Lathe, and R. E. Tanzi, The antimicrobial protection hypothesis of Alzheimer's disease, Alzheimers Dement. J. Alzheimers Assoc, vol.14, pp.1602-1614, 2018.

A. Montagne, S. R. Barnes, M. D. Sweeney, M. R. Halliday, A. P. Sagare et al., Blood-brain barrier breakdown in the aging human hippocampus, Neuron, vol.85, pp.296-302, 2015.

A. Montagne, A. M. Nikolakopoulou, Z. Zhao, A. P. Sagare, G. Si et al., Pericyte degeneration causes white matter dysfunction in the mouse central nervous system, Nat. Med, vol.24, pp.326-337, 2018.

S. Mruthinti, J. J. Buccafusco, W. D. Hill, J. L. Waller, T. W. Jackson et al., Autoimmunity in Alzheimer's disease: increased levels of circulating IgGs binding Abeta and RAGE peptides, Neurobiol. Aging, vol.25, pp.1023-1032, 2004.

M. A. Myagkova, S. I. Gavrilova, N. N. Lermontova, Y. B. Kalyn, N. D. Selezneva et al., Autoantibodies to beta-amyloid and neurotransmitters in patients with Alzheimer's disease and senile dementia of the Alzheimer type, Bull. Exp. Biol. Med, vol.131, pp.127-129, 2001.

I. A. Myles, Fast food fever: reviewing the impacts of the Western diet on immunity, Nutr. J, vol.13, p.61, 2014.

W. Na, J. Y. Shin, J. Y. Lee, S. Jeong, W. Kim et al., Dexamethasone suppresses JMJD3 gene activation via a putative negative glucocorticoid response element and maintains integrity of tight junctions in brain microvascular endothelial cells, J. Cereb. Blood Flow Metab, vol.37, pp.3695-3708, 2017.

D. A. Nation, M. D. Sweeney, A. Montagne, A. P. Sagare, L. M. Orazio et al., Regional early and progressive loss of brain pericytes but not vascular smooth muscle cells in adult mice with disrupted platelet-derived growth factor receptor-? signaling, Nat. Med, vol.25, p.176225, 2019.

S. Nishimura, I. Manabe, S. Takaki, M. Nagasaki, M. Otsu et al., Adipose natural regulatory B cells negatively control adipose tissue inflammation, Cell Metab, vol.18, pp.759-766, 2013.

F. M. Noé and N. Marchi, Central nervous system lymphatic unit, immunity, and epilepsy: is there a link?, Epilepsia Open, vol.4, pp.30-39, 2019.

R. Nortley, N. Korte, P. Izquierdo, C. Hirunpattarasilp, A. Mishra et al., Amyloid ? oligomers constrict human capillaries in Alzheimer's disease via signaling to pericytes, Science, vol.365, p.9518, 2019.

J. Ochoa-repáraz, D. W. Mielcarz, L. E. Ditrio, A. R. Burroughs, D. M. Foureau et al., Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis, J. Immunol, vol.183, pp.6041-6050, 2009.

J. Ochoa-repáraz, D. W. Mielcarz, S. Haque-begum, and L. H. Kasper, Induction of a regulatory B cell population in experimental allergic encephalomyelitis by alteration of the gut commensal microflora, Gut Microbes, vol.1, pp.103-108, 2010.

B. I. Omalu, R. L. Hamilton, M. I. Kamboh, S. T. Dekosky, and J. Bailes, Chronic traumatic encephalopathy (CTE) in a National Football League Player: case report and emerging medicolegal practice questions, J. Forensic Nurs, vol.6, pp.40-46, 2010.

A. Ouwehand, E. Isolauri, and S. Salminen, The role of the intestinal microflora for the development of the immune system in early childhood, Eur. J. Nutr, vol.41, pp.32-37, 2002.

G. Pagliai, E. Russo, E. Niccolai, M. Dinu, V. Di-pilato et al., Influence of a 3-month low-calorie Mediterranean diet compared to the vegetarian diet on human gut microbiota and SCFA: the CARDIVEG study, Eur. J. Nutr, 2019.

S. Païssé, C. Valle, F. Servant, M. Courtney, R. Burcelin et al., Comprehensive description of blood microbiome from healthy donors assessed by 16S targeted metagenomic sequencing, Transfusion, vol.56, pp.1138-1147, 2016.

F. Pani, G. Di-dalmazi, A. Corsello, T. G. Oliver, J. R. Livezey et al., MON-450 pituitary antibodies in a cohort of us service members with traumatic brain injury, J. Endocr. Soc, vol.3, p.450, 2019.

A. Parker, S. Fonseca, and S. R. Carding, Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health, Gut Microbes, 2019.

V. A. Pavlov and K. J. Tracey, Neural regulation of immunity: molecular mechanisms and clinical translation, Nat. Neurosci, vol.20, pp.156-166, 2017.

J. J. Pestka, L. L. Vines, M. A. Bates, K. He, and I. Langohr, Comparative effects of n-3, n-6 and n-9 unsaturated fatty acid-rich diet consumption on lupus nephritis, autoantibody production and CD4+ T cell-related gene responses in the autoimmune NZBWF1 mouse, PLoS One, vol.9, p.100255, 2014.

I. Petta, J. Fraussen, V. Somers, and M. Kleinewietfeld, Interrelation of diet, gut microbiome, and autoantibody production, Front. Immunol, vol.9, p.439, 2018.

S. Poole, S. K. Singhrao, S. Chukkapalli, M. Rivera, I. Velsko et al., Active invasion of Porphyromonas gingivalis and infection-induced complement activation in ApoE-/-mice brains, J. Alzheimers Dis, vol.43, pp.67-80, 2015.

E. Posse-de-chaves and S. Sipione, Sphingolipids and gangliosides of the nervous system in membrane function and dysfunction, FEBS Lett, vol.584, pp.1748-1759, 2010.

M. Prinz and J. Priller, The role of peripheral immune cells in the CNS in steady state and disease, Nat. Neurosci, vol.20, pp.136-144, 2017.

G. S. Purvis, E. Solito, and C. Thiemermann, Annexin-A1: therapeutic potential in microvascular disease, Front. Immunol, vol.10, p.938, 2019.

B. Qu, Y. Gong, C. Moore, M. Fu, D. C. German et al., Beta-amyloid auto-antibodies are reduced in Alzheimer's disease, J. Neuroimmunol, vol.274, pp.168-173, 2014.

M. Raad, E. Nohra, N. Chams, M. Itani, F. Talih et al., Autoantibodies in traumatic brain injury and central nervous system trauma, Neuroscience, vol.281, pp.16-23, 2014.

M. T. Rahman, C. Ghosh, M. Hossain, D. Linfield, F. Rezaee et al., IFN-?, IL-17A, or zonulin rapidly increase the permeability of the blood-brain and small intestinal epithelial barriers: relevance for neuroinflammatory diseases, Biochem. Biophys. Res. Commun, vol.507, pp.274-279, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02388114

R. M. Ransohoff, How neuroinflammation contributes to neurodegeneration, Science, vol.353, pp.777-783, 2016.

P. Religa, R. Cao, D. Religa, Y. Xue, N. Bogdanovic et al., VEGF significantly restores impaired memory behavior in Alzheimer's mice by improvement of vascular survival, 2013.

M. M. Reza, B. B. Finlay, and S. Pettersson, Gut microbes, ageing & organ function: a chameleon in modern biology?, EMBO Mol. Med, vol.11, p.9872, 2019.

J. O. Rinne, D. J. Brooks, M. N. Rossor, N. C. Fox, R. Bullock et al., 11C-PiB PET assessment of change in fibrillar amyloid-beta load in patients with Alzheimer's disease treated with bapineuzumab: a phase 2, doubleblind, placebo-controlled, ascending-dose study, Lancet Neurol, vol.9, pp.363-372, 2010.

H. Rosenmann, Z. Meiner, V. Geylis, O. Abramsky, and M. Steinitz, Detection of circulating antibodies against tau protein in its unphosphorylated and in its neurofibrillary tangles-related phosphorylated state in Alzheimer's disease and healthy subjects, Neurosci Lett, vol.410, pp.90-93, 2006.

S. Rudehill, S. Muhallab, A. Wennersten, C. Von-gertten, F. Al-nimer et al., Autoreactive antibodies against neurons and basal lamina found in serum following experimental brain contusion in rats, Acta Neurochir, vol.148, pp.199-205, 2006.

J. Rustenhoven, D. Jansson, L. C. Smyth, and M. Dragunow, Brain Pericytes As Mediators of Neuroinflammation, Trends Pharmacol. Sci, vol.38, pp.291-304, 2017.

S. Salloway, R. Sperling, S. Gilman, N. C. Fox, K. Blennow et al., A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease, Neurology, vol.73, pp.2061-2070, 2009.

T. R. Sampson, J. W. Debelius, T. Thron, S. Janssen, G. G. Shastri et al., Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease, Cell, vol.167, 2016.

F. Scheperjans, V. Aho, P. A. Pereira, K. Koskinen, L. Paulin et al., Gut microbiota are related to Parkinson's disease and clinical phenotype, Mov. Disord, vol.30, pp.350-358, 2015.

R. Schierwagen, C. Alvarez-silva, M. S. Madsen, C. C. Kolbe, C. Meyer et al., Circulating microbiome in blood of different circulatory compartments, 2018.

B. O. Schroeder and F. Bäckhed, Signals from the gut microbiota to distant organs in physiology and disease, Nat. Med, vol.22, pp.1079-1089, 2016.

M. Schwartz and R. Shechter, Systemic inflammatory cells fight off neurodegenerative disease, Nat. Rev. Neurol, vol.6, pp.405-410, 2010.

V. V. Senatorov, A. R. Friedman, D. Z. Milikovsky, J. Ofer, R. Saar-ashkenazy et al., Blood-brain barrier dysfunction in aging induces hyper-activation of TGF-beta signaling and chronic yet reversible neural dysfunction, Sci. Transl. Med, vol.11, p.8283, 2019.

J. Sevigny, P. Chiao, T. Bussière, P. H. Weinreb, L. Williams et al., The antibody aducanumab reduces A? plaques in Alzheimer's disease, Nature, vol.01, pp.50-56, 2016.

F. Shanahan, D. Van-sinderen, P. W. O'toole, and C. Stanton, Feeding the microbiota: transducer of nutrient signals for the host, Gut, vol.66, pp.1709-1717, 2017.

E. Sherwin, S. R. Bordenstein, J. L. Quinn, T. G. Dinan, and J. F. Cryan, Microbiota and the social brain, Science, vol.366, p.2016, 2019.

E. M. Sigurdsson, Tau immunotherapies for Alzheimer's disease and related tauopathies: progress and potential pitfalls, J. Alzheimers Dis, vol.64, pp.555-565, 2018.

R. K. Singh, H. Chang, D. Yan, K. M. Lee, D. Ucmak et al., Influence of diet on the gut microbiome and implications for human health, J. Transl. Med, vol.15, p.73, 2017.

R. K. Sinha, Y. Wang, Z. Zhao, X. Xu, L. Burnier et al., PAR1 biased signaling is required for activated protein C in vivo benefits in sepsis and stroke, Blood, vol.15, pp.1163-1171, 2018.

C. Smith, D. I. Graham, L. S. Murray, and J. A. Nicoll, Tau immunohistochemistry in acute brain injury, Neuropathol. Appl. Neurobiol, vol.29, pp.496-502, 2003.

A. Sommer, B. Winner, and I. Prots, The Trojan horseneuroinflammatory impact of T cells in neurodegenerative diseases, Mol. Neurodegener, vol.12, p.78, 2017.

E. G. Sorokina, O. M. Vol'pina, Z. B. Semenova, O. V. Karaseva, D. O. Koroev et al., , 2011.

, Zh. Nevrol. Psikhiatr. Im. S S Korsakova, vol.111, pp.56-60

S. J. Soscia, J. E. Kirby, K. J. Washicosky, S. M. Tucker, M. Ingelsson et al., The Alzheimer's disease-associated amyloid beta-protein is an antimicrobial peptide, PLoS One, vol.5, p.9505, 2010.

E. J. Su, L. Fredriksson, M. Kanzawa, S. Moore, E. Folestad et al., Imatinib treatment reduces brain injury in a murine model of traumatic brain injury, Front. Cell. Neurosci, vol.9, p.385, 2015.

M. D. Sweeney, Z. Zhao, A. Montagne, A. R. Nelson, and B. V. Zlokovic, Blood-brain barrier: from physiology to disease and back, Physiol. Rev, vol.01, pp.21-78, 2019.

X. Y. Tai, M. Koepp, J. S. Duncan, N. Fox, P. Thompson et al., Hyperphosphorylated tau in patients with refractory epilepsy correlates with cognitive decline: a study of temporal lobe resections, Brain J. Neurol, vol.139, pp.2441-2455, 2016.

J. Tanaka, K. Nakamura, M. Takeda, K. Tada, H. Suzuki et al., Enzyme-linked immunosorbent assay for human autoantibody to glial fibrillary acidic protein: higher titer of the antibody is detected in serum of patients with Alzheimer's disease, Acta Neurol. Scand, vol.80, pp.554-560, 1989.

F. Tanriverdi, A. De-bellis, M. Battaglia, G. Bellastella, A. Bizzarro et al., Investigation of antihypothalamus and antipituitary antibodies in amateur boxers: is chronic repetitive head trauma-induced pituitary dysfunction associated with autoimmunity?, Eur. J. Endocrinol, vol.162, pp.861-867, 2010.

F. Tanriverdi, A. De-bellis, A. Bizzarro, A. A. Sinisi, G. Bellastella et al., Antipituitary antibodies after traumatic brain injury: is head trauma-induced pituitary dysfunction associated with autoimmunity?, Eur. J. Endocrinol, vol.159, pp.7-13, 2008.

F. Tanriverdi, H. Senyurek, K. Unluhizarci, A. Selcuklu, F. F. Casanueva et al., High risk of hypopituitarism after traumatic brain injury: a prospective investigation of anterior pituitary function in the acute phase and 12 months after trauma, J. Clin. Endocrinol. Metab, vol.91, pp.2105-2111, 2006.

M. Thiyagarajan, J. A. Fernández, S. M. Lane, J. H. Griffin, and B. V. Zlokovic, Activated protein C promotes neovascularization and neurogenesis in postischemic brain via protease-activated receptor 1, J. Neurosci, vol.28, pp.12788-12797, 2008.

T. Thomas, G. Thomas, C. Mclendon, T. Sutton, and M. Mullan, beta-Amyloid-mediated vasoactivity and vascular endothelial damage, Nature, vol.380, pp.168-171, 1996.

V. Tomasdottir, S. Thorleifsdottir, A. Vikingsson, I. Hardardottir, and J. Freysdottir, Dietary omega-3 fatty acids enhance the B1 but not the B2 cell immune response in mice with antigen-induced peritonitis, J. Nutr. Biochem, vol.25, pp.111-117, 2014.

, A Long-Term Safety and Tolerability Extension Study Of Bapineuzumab In Alzheimer Disease Patients, 2019.

, A Long-Term Safety and Tolerability Study Of Bapineuzumab In Alzheimer Disease Patients, 2019.

D. Vacirca, F. Delunardo, P. Matarrese, T. Colasanti, P. Margutti et al., Autoantibodies to the adenosine triphosphate synthase play a pathogenetic role in Alzheimer's disease, Neurobiol. Aging, vol.33, pp.753-766, 2012.

C. E. Van-skike, J. B. Jahrling, A. B. Olson, N. L. Sayre, S. A. Hussong et al., Inhibition of mTOR protects the blood-brain barrier in models of Alzheimer's disease and vascular cognitive impairment, Am. J. Physiol. Heart Circ. Physiol, vol.314, pp.693-703, 2018.

A. Vezzani, J. French, T. Bartfai, and T. Z. Baram, The role of inflammation in epilepsy, Nat. Rev. Neurol, vol.7, pp.31-40, 2011.

A. T. Virtue, S. J. Mccright, J. M. Wright, M. T. Jimenez, W. K. Mowel et al., The gut microbiota regulates white adipose tissue inflammation and obesity via a family of microRNAs, Sci. Transl. Med, vol.11, p.1892, 2019.

N. M. Vogt, R. L. Kerby, K. A. Dill-mcfarland, S. J. Harding, A. P. Merluzzi et al., Gut microbiome alterations in Alzheimer's disease, Sci. Rep, vol.7, p.13537, 2017.

X. Wang, G. Sun, T. Feng, J. Zhang, X. Huang et al., Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer's disease progression, Cell Res, vol.29, pp.787-803, 2019.

Y. Wang, Z. Zhao, S. V. Rege, M. Wang, G. Si et al., 3K3A-activated protein C stimulates postischemic neuronal repair by human neural stem cells in mice, Nat. Med, vol.22, pp.1050-1055, 2016.

M. E. Weksler, N. Relkin, R. Turkenich, S. Larusse, L. Zhou et al., Patients with Alzheimer disease have lower levels of serum anti-amyloid peptide antibodies than healthy elderly individuals, Exp. Gerontol, vol.37, pp.943-948, 2002.

J. S. Wilson, S. Vainti, J. J. Buccafusco, R. F. Schade, M. B. Mitchell et al., Anti-RAGE and Abeta immunoglobulin levels are related to dementia level and cognitive performance, J. Gerontol. A Biol. Sci. Med. Sci, vol.64, pp.264-271, 2009.

J. Wu, L. , and L. , Autoantibodies in Alzheimer's disease: potential biomarkers, pathogenic roles, and therapeutic implications, J. Biomed. Res, vol.30, pp.361-372, 2016.

F. Zhang, C. Eckman, S. Younkin, K. K. Hsiao, and C. Iadecola, Increased susceptibility to ischemic brain damage in transgenic mice overexpressing the amyloid precursor protein, J. Neurosci, vol.17, pp.7655-7661, 1997.

N. Zhang, S. Yin, W. Zhang, X. Gong, N. Zhang et al., Crystal structure and biochemical characterization of an aminopeptidase LapB from Legionella pneumophila, J. Agric. Food Chem, vol.65, pp.7569-7578, 2017.

Z. Zhang, J. S. Zoltewicz, S. Mondello, K. J. Newsom, Z. Yang et al., Human traumatic brain injury induces autoantibody response against glial fibrillary acidic protein and its breakdown products, PLoS One, vol.9, p.92698, 2014.

P. Zheng, B. Zeng, C. Zhou, M. Liu, Z. Fang et al., Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host's metabolism, Mol. Psychiatry, vol.21, pp.786-796, 2016.

Z. Zhong, H. Ilieva, L. Hallagan, R. Bell, I. Singh et al., Activated protein C therapy slows ALS-like disease in mice by transcriptionally inhibiting SOD1 in motor neurons and microglia cells, J. Clin. Invest, vol.119, pp.3437-3449, 2009.

Z. Zhuang, L. Shen, W. Li, X. Fu, F. Zeng et al., Gut microbiota is altered in patients with Alzheimer's disease, J. Alzheimers Dis, vol.63, pp.1337-1346, 2018.

B. V. Zlokovic, The blood-brain barrier in health and chronic neurodegenerative disorders, Neuron, vol.57, pp.178-201, 2008.

B. V. Zlokovic, Neurovascular pathways to neurodegeneration in Alzheimer's disease and other disorders, Nat. Rev. Neurosci, vol.12, pp.723-738, 2011.

E. Zub, G. Canet, R. Garbelli, M. Blaquiere, L. Rossini et al., The GR-ANXA1 pathway is a pathological player and a candidate target in epilepsy, FASEB J, vol.33, pp.13998-14009, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02350122

. Copyright-©-2020-giannoni, N. Claeysen, and M. , This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited