L. Abuqayyas and J. P. Balthasar, Investigation of the role of FcR and FcRn in mAb distribution to the brain, Mol Pharm, vol.10, pp.1505-1518, 2013.

B. Allen, E. Ingram, M. Takao, M. J. Smith, R. Jakes et al., Abundant tau filaments and nonapoptotic neurodegeneration in transgenic mice expressing human P301S tau protein, J Neurosci, vol.22, pp.9340-51, 2002.

A. C. Alonso, I. Grundke-iqbal, and K. Iqbal, Alzheimer's disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules, Nat Med, vol.2, pp.783-790, 1996.

Z. Ahmed, J. Cooper, T. K. Murray, K. Garn, E. Mcnaughton et al., A novel in vivo model of tau propagation with rapid and progressive neurofibrillary tangle pathology: the pattern of spread is determined by connectivity, not proximity, Acta Neuropathol, vol.127, pp.667-83, 2014.

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.9115-9144, 2007.

J. C. Augustinack, A. Schneider, E. M. Mandelkow, and B. T. Hyman, Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer's disease, Acta Neuropathol, vol.103, pp.26-35, 2002.

N. R. Barthélemy, F. Fenaille, C. Hirtz, N. Sergeant, S. Schraen-maschke et al., Tau protein quantification in human cerebrospinal fluid by targeted mass spectrometry at high sequence coverage provides insights into its primary structure heterogeneity, J Proteome Res, vol.15, pp.667-76, 2016.

N. R. Barthélemy, A. Gabelle, C. Hirtz, F. Fenaille, N. Sergeant et al., Differential mass spectrometry profiles of tau protein in the cerebrospinal fluid of patients with Alzheimer's disease, progressive supranuclear palsy, and dementia with lewy bodies, J Alzheimers Dis, vol.51, pp.1033-1076, 2016.

A. Boutajangout, J. Ingadottir, P. Davies, and E. M. Sigurdsson, Passive immunization targeting pathological phospho-tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain, J Neurochem, vol.118, pp.658-67, 2011.

H. Braak and E. Braak, Neuropathological stageing of Alzheimer-related changes, Acta Neuropathol, vol.82, pp.239-59, 1991.

H. Braak, D. Tredici, and K. , Alzheimer's pathogenesis: is there neuron-toneuron propagation?, Acta Neuropathol, vol.121, pp.589-95, 2011.

L. Buée, T. Bussière, V. Buée-scherrer, A. Delacourte, and P. R. Hof, Tau protein isoforms, phosphorylation and role in neurodegenerative disorders, Brain Res Brain Res Rev, vol.33, pp.95-130, 2000.

V. Buée-scherrer, O. Condamines, C. Mourton-gilles, R. Jakes, M. Goedert et al., AD2, a phosphorylation-dependent monoclonal antibody directed against tau proteins found in Alzheimer's disease, Brain Res Mol Brain Res, vol.39, pp.79-88, 1996.

S. Calafate, A. Buist, K. Miskiewicz, V. Vijayan, G. Daneels et al., Synaptic contacts enhance cell-to-cell tau pathology propagation, Cell Rep, vol.11, pp.1176-83, 2015.

D. L. Castillo-carranza, M. J. Guerrero-muñ-oz, U. Sengupta, C. Hernandez, A. D. Barrett et al., Tau immunotherapy modulates both pathological tau and upstream amyloid pathology in an Alzheimer's disease mouse model, J Neurosci, vol.35, pp.4857-68, 2015.

D. L. Castillo-carranza, U. Sengupta, M. J. Guerrero-muñ-oz, C. A. Lasagna-reeves, J. E. Gerson et al., Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles, J Neurosci, vol.34, pp.4260-72, 2014.

X. Chai, S. Wu, T. K. Murray, R. Kinley, C. V. Cella et al., Passive immunization with anti-Tau antibodies in two transgenic models: reduction of Tau pathology and delay of disease progression, J Biol Chem, vol.286, pp.34457-67, 2011.

H. Cho, J. Y. Choi, M. S. Hwang, Y. J. Kim, H. M. Lee et al., In vivo cortical spreading pattern of tau and amyloid in the Alzheimer disease spectrum, Ann Neurol, vol.80, pp.247-58, 2016.

F. Clavaguera, T. Bolmont, R. A. Crowther, D. Abramowski, S. Frank et al., Transmission and spreading of tauopathy in transgenic mouse brain, Nat Cell Biol, vol.11, pp.909-922, 2009.

L. Collin, B. Bohrmann, U. Gö-pfert, K. Oroszlan-szovik, and L. Ozmen, Grü ninger F. Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimer's disease, Brain, vol.137, pp.2834-2880, 2014.

E. E. Congdon, Y. Lin, H. B. Rajamohamedsait, D. B. Shamir, S. Krishnaswamy et al., Affinity of Tau antibodies for solubilized pathological Tau species but not their immunogen or insoluble Tau aggregates predicts in vivo and ex vivo efficacy, Mol Neurodegener, vol.11, p.62, 2016.

T. E. Cope, T. Rittman, R. J. Borchert, P. S. Jones, D. Vatansever et al., Tau burden and the functional connectome in Alzheimer's disease and progressive supranuclear palsy, Brain, vol.141, pp.550-67, 2018.

J. P. Courade, G. Mairet-coello, . Tyson-k-b, and D. Lightwood, Epitope determines efficacy of therapeutic anti-Tau antibodies in a functional assay with human Alzheimer Tau, Acta Neuropathol, vol.136, pp.729-774, 2018.

C. L. Croft, M. A. Wade, K. Kurbatskaya, P. Mastrandreas, M. M. Hughes et al., Membrane association and release of wild-type and pathological tau from organotypic brain slice cultures, Cell Death Dis, vol.8, p.2671, 2017.

C. D'abramo, C. M. Acker, H. T. Jimenez, and P. Davies, Tau passive immunotherapy in mutant P301L mice: antibody affinity versus specificity, PLoS One, vol.8, p.62402, 2013.

C. L. Dai, X. Chen, S. F. Kazim, F. Liu, C. X. Gong et al., Passive immunization targeting the N-terminal projection domain of tau decreases tau pathology and improves cognition in a transgenic mouse model of Alzheimer disease and tauopathies, J Neural Transm, vol.122, pp.607-624, 2015.

C. L. Dai, Y. C. Tung, F. Liu, C. X. Gong, and K. Iqbal, Tau passive immunization inhibits not only tau but also Ab pathology, Alzheimers Res Ther, vol.9, p.1, 2017.

C. L. Dai, W. Hu, Y. C. Tung, F. Liu, C. X. Gong et al., Tau passive immunization blocks seeding and spread of Alzheimer hyperphosphorylated Tau-induced pathology in 3 Â Tg-AD mice, Alzheimers Res Ther, vol.10, p.13, 2018.

A. De-calignon, M. Polydoro, M. Suá-rez-calvet, C. William, D. H. Adamowicz et al., Propagation of tau pathology in a model of early Alzheimer's disease, Neuron, vol.73, pp.685-97, 2012.

A. Delacourte, J. P. David, N. Sergeant, L. Buée, A. Wattez et al., The biochemical pathway of neurofibrillary degeneration in aging and Alzheimer's disease, Neurology, vol.52, pp.1158-65, 1999.

M. Derisbourg, C. Leghay, G. Chiappetta, F. J. Fernandez-gomez, C. Laurent et al., Role of the Tau N-terminal region in microtubule stabilization revealed by new endogenous truncated forms, Sci Rep, vol.5, p.9659, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01659458

S. Dujardin, K. Lécolle, R. Caillierez, S. Bégard, N. Zommer et al., Neuron-to-neuron wild-type Tau protein transfer through a trans-synaptic mechanism: relevance to sporadic tauopathies, Acta Neuropathol Commun, vol.2, p.14, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-00945726

S. Dujardin, S. Begard, R. Caillierez, C. Lachaud, L. Delattre et al., Ectosomes: a new mechanism for non-exosomal secretion of tau protein, PLoS ONE, vol.9, p.100760, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01181185

C. Duyckaerts, M. Bennecib, Y. Grignon, T. Uchihara, Y. He et al., Modeling the relation between neurofibrillary tangles and intellectual status, Neurobiol Aging, vol.18, pp.267-73, 1997.

C. Duyckaerts, D. Seilhean, V. Sazdovitch, I. Plu, B. Delatour et al., Seeding and propagation of lesions in neurodegenerative diseases: a new paradigm, Bull Acad Natl Med, vol.199, pp.809-828, 2015.

M. D'orange, G. Aurégan, D. Cheramy, M. Gaudin-guérif, S. Lieger et al., Potentiating tangle formation reduces acute toxicity of soluble tau species in the rat, Brain, vol.141, pp.535-584, 2018.

S. K. Forest, C. M. Acker, C. Abramo, and P. Davies, Methods for measuring tau pathology in transgenic mouse models, J Alzheimers Dis, vol.33, pp.463-71, 2013.

K. E. Funk, H. Mirbaha, H. Jiang, D. M. Holtzman, and M. I. Diamond, Distinct therapeutic mechanisms of tau antibodies: promoting microglial clearance versus blocking neuronal uptake, J Biol Chem, vol.290, pp.21652-62, 2015.

M. Goedert, M. G. Spillantini, R. Jakes, D. Rutherford, and R. A. Crowther, Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer's disease, Neuron, vol.3, pp.519-545, 1989.

D. P. Hanger, D. H. Lau, E. C. Phillips, M. K. Bondulich, T. Guo et al., Intracellular and extracellular roles for tau in neurodegenerative disease, J Alzheimers Dis, vol.40, issue.1, pp.37-45, 2014.

M. C. Hoenig, G. N. Bischof, J. Seemiller, J. Hammes, J. Kukolja et al., Networks of tau distribution in Alzheimer's disease, Brain, vol.14, pp.568-81, 2018.

J. J. Iliff, M. J. Chen, B. A. Plog, D. M. Zeppenfeld, M. Soltero et al., Impairment of glymphatic pathway function promotes tau pathology after traumatic brain injury, J Neurosci, vol.34, pp.16180-93, 2014.

S. Jadhav, J. Avila, M. Schö-ll, G. G. Kovacs, E. Kö-vari et al., A walk through tau therapeutic strategies, Acta Neuropathol Commun, vol.7, p.22, 2019.

G. A. Jicha, R. Bowser, I. G. Kazam, and P. Davies, Alz-50 and MC-1, a new monoclonal antibody raised to paired helical filaments, recognize conformational epitopes on recombinant tau, J Neurosci Res, vol.48, pp.128-160, 1997.

K. Leroy, A. Bretteville, K. Schindowski, E. Gilissen, M. Authelet et al., Early axonopathy preceding neurofibrillary tangles in mutant tau transgenic mice, Am J Pathol, vol.171, pp.976-92, 2007.

A. Mudher, M. Colin, S. Dujardin, M. Medina, I. Dewachter et al., What is the evidence that tau pathology spreads through prion-like propagation?, Acta Neuropathol Commun, vol.5, p.99, 2017.

P. Nacharaju, J. Lewis, C. Easson, Y. S. Hackett, J. Hutton et al., Accelerated filament formation from tau protein with specific FTDP-17 missense mutations, FEBS Lett, vol.447, pp.195-204, 1999.

C. K. Nobuhara, S. L. Devos, C. Commins, S. Wegmann, B. D. Moore et al., Tau antibody targeting pathological species blocks neuronal uptake and interneuron propagation of tau in Vitro, Am J Pathol, vol.187, pp.1399-412, 2017.

J. T. Pedersen and E. M. Sigurdsson, Tau immunotherapy for Alzheimer's disease, Trends Mol Med, vol.21, pp.394-402, 2015.

E. Peeraer, A. Bottelbergs, K. Van-kolen, I. C. Stancu, B. Vasconcelos et al., Intracerebral injection of preformed synthetic tau fibrils initiates widespread tauopathy and neuronal loss in the brains of tau transgenic mice, Neurobiol Dis, vol.73, pp.83-95, 2015.

Y. Saito, N. N. Ruberu, M. Sawabe, T. Arai, N. Tanaka et al., Staging of argyrophilic grains: an age-associated tauopathy, J Neuropathol Exp Neurol, vol.63, pp.911-919, 2004.

S. Sankaranarayanan, D. M. Barten, L. Vana, N. Devidze, L. Yang et al., Passive immunization with phospho-tau antibodies reduces tau pathology and functional deficits in two distinct mouse tauopathy models, PLoS One, vol.10, p.125614, 2015.

C. Sato, N. R. Barthélemy, K. G. Mawuenyega, B. W. Patterson, B. A. Gordon et al., Tau Kinetics in neurons and the human central nervous system, Neuron, vol.98, pp.861-64, 2018.

K. Schindowski, A. Bretteville, K. Leroy, S. Bégard, J. P. Brion et al., Alzheimer's disease-like tau neuropathology leads to memory deficits and loss of functional synapses in a novel mutated tau transgenic mouse without any motor deficits, Am J Pathol, vol.169, pp.599-616, 2006.

I. Sotiropoulos, M. C. Galas, J. M. Silva, E. Skoulakis, S. Wegmann et al., 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-01654098

M. G. Spillantini, T. D. Bird, and B. Ghetti, Frontotemporal dementia and parkinsonism linked to chromosome 17: a new group of tauopathies, Brain Pathol, vol.8, pp.387-402, 1998.

I. St-amour, I. Paré, W. Alata, K. Coulombe, C. Ringuette-goulet et al., Brain bioavailability of human intravenous immunoglobulin and its transport through the murine blood-brain barrier, J Cereb Blood Flow Metab, vol.33, pp.1983-92, 2013.

J. H. Su, G. Deng, and C. W. Cotman, Transneuronal degeneration in the spread of Alzheimer's disease pathology: immunohistochemical evidence for the transmission of tau hyperphosphorylation, Neurobiol Dis, vol.4, pp.365-75, 1997.

D. Terwel, R. Lasrado, J. Snauwaert, E. Vandeweert, C. Van-haesendonck et al., Changed conformation of mutant Tau-P301L underlies the moribund tauopathy, absent in progressive, nonlethal axonopathy of Tau-4R/2N transgenic mice, J Biol Chem, vol.280, pp.3963-73, 2005.

L. Troquier, R. Caillierez, S. Burnouf, F. J. Fernandez-gomez, M. E. Grosjean et al., Targeting phospho-Ser422 by active Tau Immunotherapy in the THYTau22 mouse model: a suitable therapeutic approach, Curr Alzheimer Res, vol.9, pp.397-405, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00664452

T. Umeda, H. Eguchi, Y. Kunori, Y. Matsumoto, T. Taniguchi et al., Passive immunotherapy of tauopathy targeting pSer413-tau: a pilot study in mice, Ann Clin Transl Neurol, vol.2, pp.241-55, 2015.

M. Vandermeeren, M. Borgers, K. Van-kolen, C. Theunis, B. Vasconcelos et al., Anti-Tau monoclonal antibodies derived from soluble and filamentous tau show diverse functional properties in vitro and in vivo, J Alzheimers Dis, vol.65, pp.265-81, 2018.

M. Verny, C. Duyckaerts, Y. Agid, and J. J. Hauw, The significance of cortical pathology in progressive supranuclear palsy. Clinico-pathological data in 10 cases, Brain, vol.119, pp.1123-1159, 1996.

A. Watanabe, K. Takio, and Y. Ihara, Deamidation and isoaspartate formation in smeared tau in paired helical filaments. Unusual properties of the microtubule-binding domain of tau, J Biol Chem, vol.274, pp.7368-78, 1999.

M. D. Weingarten, A. H. Lockwood, S. Y. Hwo, and M. W. Kirschner, A protein factor essential for microtubule assembly, Proc Natl Acad Sci, vol.72, pp.1858-62, 1975.

D. R. Williams, J. L. Holton, C. Strand, A. Pittman, R. De-silva et al., Pathological tau burden and distribution distinguishes progressive supranuclear palsy-parkinsonism from Richardson's syndrome, Brain, vol.130, pp.1566-76, 2007.

C. M. Wischik, M. Novak, H. C. Thøgersen, P. C. Edwards, M. J. Runswick et al., Isolation of a fragment of tau derived from the core of the paired helical filament of Alzheimer disease, Proc Natl Acad Sci, vol.85, pp.4506-4516, 1988.

K. Yanamandra, T. K. Patel, H. Jiang, S. Schindler, J. D. Ulrich et al., Anti-tau antibody administration increases plasma tau in transgenic mice and patients with tauopathy, Sci Transl Med, vol.9, 2017.

N. Zilka, B. Kovacech, P. Barath, E. Kontsekova, and M. Novak, The selfperpetuating tau truncation circle, Biochem Soc Trans, vol.40, pp.681-687, 2012.