S. Hannaoui, A. Gougerot, N. Privat, and E. Levavasseur,

M. D. Adams, S. E. Celniker, R. A. Holt, C. A. Evans, J. D. Gocayne et al., The genome sequence of Drosophila melanogaster, Science, vol.287, pp.2185-95, 2000.

K. T. Adjou, R. Demaimay, C. Lasmezas, J. P. Deslys, M. Seman et al., MS-8209, a new amphotericin B derivative, provides enhanced efficacy in delaying hamster scrapie, Antimicrob Agents Chemother, vol.39, pp.2810-2822, 1995.

K. T. Adjou, J. P. Deslys, R. Demaimay, and D. Dormont, Probing the dynamics of prion diseases with amphotericin B, Trends Microbiol, vol.5, pp.27-31, 1997.

K. T. Adjou, N. Privat, S. Demart, J. P. Deslys, M. Seman et al., MS-8209, an amphotericin B analogue, delays the appearance of spongiosis, astrogliosis and PrPres accumulation in the brain of scrapie-infected hamsters, J Comp Pathol, vol.122, pp.3-8, 2000.

K. T. Adjou, S. Simoneau, N. Sales, F. Lamoury, D. Dormont et al., A novel generation of heparan sulfate mimetics for the treatment of prion diseases, J Gen Virol, vol.84, pp.2595-603, 2003.

Y. Aguib, A. Heiseke, S. Gilch, C. Riemer, M. Baier et al., Autophagy induction by trehalose counter-acts cellular prioninfection, Autophagy, vol.5, pp.361-69, 2009.

A. Aguzzi, F. Baumann, and J. Bremer, The prion's elusive reason for being, Annu Rev Neurosci, vol.31, pp.439-77, 2008.

A. Alzualde, B. Indakoetxea, I. Ferrer, F. Moreno, M. Barandiaran et al., A Novel PRNP Y218N Mutation in Gerstmann-Straüssler-Scheinker Disease With Neurofibrillary Degeneration, J Neuropathol Exp Neurol, vol.69, pp.789-800, 2010.

C. Andorfer, Y. Kress, M. Espinoza, R. Silva, K. L. Tucker et al., Hyperphosphorylation and aggregation of tau in mice expressing normal human tau isoforms, Journal of Neurochemistry, vol.86, pp.582-90, 2003.

R. Andre and S. J. Tabrizi, Misfolded PrP and a novel mechanism of proteasome inhibition, Prion, vol.6, pp.32-38, 2012.

O. Andreoletti, N. Morel, C. Lacroux, V. Rouillon, C. Barc et al., Bovine spongiform encephalopathy agent in spleen from an ARR/ARR orally exposed sheep, J Gen Virol, vol.87, pp.1043-1049, 2006.

F. Archer, C. Bachelin, O. Andreoletti, N. Besnard, G. Perrot et al., Cultured Peripheral Neuroglial Cells Are Highly Permissive to Sheep Prion Infection, Journal of Virology, vol.78, pp.482-90, 2003.

T. Arendt, J. Stieler, A. M. Strijkstra, R. A. Hut, J. Rüdiger et al., Reversible paired helical filament-like phosphorylation of tau is an adaptive process associated with neuronal plasticity in hibernating animals, J Neurosci, vol.23, pp.6972-81, 2003.

P. V. Arriagada, J. H. Growdon, E. T. Hedley-whyte, and B. T. Hyman, Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer's disease, Neurology, vol.42, pp.631-670, 1992.

E. A. Asante, I. Gowland, A. Grimshaw, J. M. Linehan, M. Smidak et al., Absence of spontaneous disease and comparative prion susceptibility of transgenic mice expressing mutant human prion proteins, J Gen Virol, vol.90, pp.546-58, 2009.

E. A. Asante, J. M. Linehan, M. Smidak, A. Tomlinson, A. Grimshaw et al., Inherited prion disease A117V is not simply a proteinopathy but produces prions transmissible to transgenic mice expressing homologous prion protein, PLoS Pathog, vol.9, p.1003643, 2013.

R. Atarashi, K. Satoh, K. Sano, T. Fuse, N. Yamaguchi et al.,

N. Collins and . Nishida, Ultrasensitive human prion detection in cerebrospinal fluid by real-time quaking-induced conversion, Nat Med, vol.17, pp.175-183, 2011.

R. Atarashi, J. M. Wilham, L. Christensen, A. G. Hughson, R. A. Moore et al., Simplified ultrasensitive prion detection by recombinant PrP conversion with shaking, Nat Med, vol.5, pp.211-223, 2008.

J. D. Atkin, M. A. Farg, A. K. Walker, C. Mclean, D. Tomas et al., Endoplasmic reticulum stress and induction of the unfolded protein response in human sporadic amyotrophic lateral sclerosis, Neurobiol Dis, vol.30, pp.400-407, 2008.

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.

G. S. Baron, A. C. Magalhaes, M. A. Prado, and B. Caughey, Mouse-adapted scrapie infection of SN56 cells: greater efficiency with microsome-associated versus purified PrP-res, J Virol, vol.80, pp.2106-2123, 2006.

A. Barret, F. Tagliavini, G. Forloni, C. Bate, M. Salmona et al., Evaluation of Quinacrine Treatment for Prion Diseases, Journal of Virology, vol.77, pp.8462-69, 2003.

I. V. Baskakov, Autocatalytic conversion of recombinant prion proteins displays a species barrier, J Biol Chem, vol.279, pp.7671-7678, 2004.

I. V. Baskakov, G. Legname, M. A. Baldwin, S. B. Prusiner, and F. E. Cohen, Pathway complexity of prion protein assembly into amyloid, J Biol Chem, vol.277, pp.21140-21148, 2002.

K. Basler, B. Oesch, M. Scott, D. Westaway, M. Wälchli et al., Scrapie and Cellular PrP Isoforms Are Encoded by the Same Chromosomal Gene, Cell, vol.46, pp.417-445, 1986.

C. Bate, S. Reid, and A. Williams, Killing of prion-damaged neurones by microglia, Neuroreport, vol.12, pp.2589-94, 2001.

C. Bate and A. Williams, Amyloid-beta-induced synapse damage is mediated via cross-linkage of cellular prion proteins, J Biol Chem, vol.286, pp.37955-63, 2011.

M. J. Bautista, J. Gutierrez, F. J. Salguero, M. M. Fernandez-de-marco, J. L. Romero-trevejo et al., BSE infection in bovine PrP transgenic mice leads to hyperphosphorylation of tau-protein, Vet Microbiol, vol.115, pp.293-301, 2006.

U. Baxa, R. B. Wickner, A. C. Steven, D. E. Anderson, L. N. Marekov et al., Characterization of beta-sheet structure in Ure2p1-89 yeast prion fibrils by solid-state nuclear magnetic resonance, Biochem, vol.46, pp.13149-62, 2007.

M. Baylis and W. Goldmann, The genetics of scrapie in sheep and goats', Curr Mol Med, vol.4, pp.385-96, 2004.

J. F. Bazan, R. J. Fletterick, M. P. Mckinley, and S. B. Prusiner, Predicted secondary structure and membrane topology of the scrapie prion protein, Protein Eng, vol.1, pp.125-160, 1987.

R. Walker, E. G. Mc, and . Geer, Patterns of gliosis in Alzheimer's disease and aging cerebrum, Glia, vol.2, pp.420-456, 1989.

S. L. Benestad, P. Sarradin, B. Thu, J. Schönheit, M. A. Tranulis et al., Cases of scrapie with unusual features in Norway and designation of a new type, Vet Rec, vol.153, pp.202-210, 2003.

J. Benito-león, Combined quinacrine and chlorpromazine therapy in fatal familial insomnia, Clin Neuropharmacol, vol.27, pp.201-204, 2004.

V. Béringue, L. Herzog, E. Jaumain, F. Reine, P. Sibille et al., Facilitated cross-species transmission of prions in extraneural tissue, Science, vol.335, pp.472-75, 2012.

V. Beringue, A. L. Dur, P. Tixador, F. Reine, L. Lepourry et al., Prominent and persistent extraneural infection in human PrP transgenic mice infected with variant CJD, PLoS One, vol.3, p.1419, 2008.

V. Beringue, D. Vilette, G. Mallinson, F. Archer, M. Kaisar et al., PrPSc binding antibodies are potent inhibitors of prion replication in cell lines, J Biol Chem, vol.279, pp.39671-39677, 2004.

D. B. Berry, D. Lu, M. Geva, J. C. Watts, S. Bhardwaj et al., Drug resistance confounding prion therapeutics, Proc Natl Acad Sci U S A, vol.110, pp.4160-69, 2013.

R. A. Bessen and R. F. Marsh, Distinct PrP Properties Suggest the Molecular Basis of Strain Variation in Transmissible Mink Encephalopathy, J Gen Virol, vol.73, pp.7859-68, 1992.

R. A. Bessen, H. Shearin, S. Martinka, R. Boharski, D. Lowe et al., Prion shedding from olfactory neurons into nasal secretions, PLoS Pathog, vol.6, p.1000837, 2010.

J. Biernat, N. Gustke, G. Drewes, E. M. Mandelkow, and E. Mandelkow, Phosphorylation of Ser262 strongly reduces binding of tau to microtubules: distinction between PHF-like immunoreactivity and microtubule binding, Neuron, vol.11, pp.153-63, 1993.

J. Biernat and E. M. Mandelkow, The development of cell processes induced by tau protein requires phosphorylation of serine 262 and 356 in the repeat domain and is inhibited by phosphorylation in the proline-rich domains, Mol Biol Cell, vol.10, pp.727-767, 1999.

J. Biernat, Y. Z. Wu, T. Timm, Q. Zheng-fischhofer, E. Mandelkow et al., Protein kinase MARK/PAR-1 is required for neurite outgrowth and establishment of neuronal polarity, Mol Biol Cell, vol.13, pp.4013-4041, 2002.

M. T. Bishop, R. G. Will, and J. C. Manson, Defining sporadic Creutzfeldt-Jakob disease strains and their transmission properties, Proc Natl Acad Sci U S A, vol.107, pp.12005-12015, 2010.

N. Bizat, J. M. Peyrin, S. Haïk, V. Cochois, P. Beaudry et al., Neuron dysfunction is induced by prion protein with an insertional mutation via a Fyn kinase and reversed by sirtuin activation in Caenorhabditis elegans, J Neurosci, vol.30, pp.5394-403, 2010.

O. V. Bocharova, L. Breydo, A. S. Parfenov, V. V. Salnikov, and I. V. Baskakov, In vitro conversion of full-length mammalian prion protein produces amyloid form with physical properties of PrP(Sc), J Mol Biol, vol.346, pp.645-59, 2005.

P. J. Bosque and S. B. Prusiner, Cultured Cell Sublines Highly Susceptible to Prion Infection, J Virol, vol.74, pp.4377-86, 2000.

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

J. P. Brandel, C. A. Heath, M. W. Head, E. Levavasseur, R. Knight et al., Variant Creutzfeldt-Jakob disease in France and the United Kingdom: Evidence for the same agent strain, Ann Neurol, vol.65, pp.249-56, 2009.

J. P. Brandel, L. Peckeu, and S. Haïk, The French surveillance network of Creutzfeldt-Jakob disease. Epidemiological data in France and worldwide, Transfus Clin Biol, vol.20, pp.395-402, 2013.

J. Bremer, F. Baumann, C. Tiberi, C. Wessig, H. Fischer et al., Axonal prion protein is required for peripheral myelin maintenance, Nat Neurosci, vol.13, pp.310-318, 2010.

S. Brenner, THE GENETICS OF CAENORHABDITIS ELEGANS, vol.77, pp.71-94, 1974.

D. R. Brown, Copper and prion disease, Brain Res Bull, vol.55, pp.165-73, 2001.

K. L. Brown, J. Brown, D. L. Ritchie, N. Sales, and J. R. Fraser, Fetal cell grafts provide longterm protection against scrapie induced neuronal loss, Neuroreport, vol.12, pp.77-82, 2001.

M. E. Bruce, I. Mcconnell, H. Fraser, and A. G. Dickinson, The disease characteristics of differentstrains of scrapie in Sinc congenic mouse lines: implications for the nature of the agent and host control of pathogenesis, J Gen Virol, vol.72, pp.595-603, 1991.

M. E. Bruce, P. A. Mcbride, and C. F. Farquhar, Precise targeting of the pathology of the sialoglycoprotein, PrP, and vacuolar degeneration in mouse scrapie, Neurosci Lett, vol.102, pp.1-6, 1989.

M. E. Bruce, R. G. Will, J. W. Ironside, I. Mcconnell, D. Drummond et al., Transmissions to mice indicate that 'new variant' CJD is caused by the BSE agent, Nature, vol.389, pp.498-501, 1997.

C. J. Buchholz, P. Bach, D. Nikles, and U. Kalinke, Prion protein-specific antibodies for therapeutic intervention of transmissible spongiform encephalopathies, Expert Opin Biol Ther, vol.6, pp.293-300, 2006.

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.

H. Büeler, A. Aguzzi, A. Saller, R. Greiner, P. Autenried et al., Mice devoid of PrP are resistant to scrapie, Cell, vol.73, pp.1339-1386, 1993.

H. Büeler, M. Fischer, Y. Lang, H. Bluethmann, H. P. Lipp et al., Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein, Nature, vol.356, pp.577-82, 1992.

A. Buschmann, G. Luhken, J. Schultz, G. Erhardt, and M. H. Groschup, Neuronal accumulation of abnormal prion protein in sheep carrying a scrapie-resistant genotype (PrPARR/ARR), J Gen Virol, vol.85, pp.2727-2760, 2004.

A. Buschmann, E. Pfaff, K. Reifenberg, H. M. Müller, and M. H. Groschup, Detection of cattle-derived BSE prions using transgenic mice overexpressing bovine PrP(C), Arch Virol Suppl, vol.16, pp.75-86, 2000.

D. A. Butler, M. R. Scott, J. M. Bockman, D. R. Borchelt, A. Taraboulos et al., Scrapie-Infected Murine Neuroblastoma Cells Produce Protease-Resistant Prion Proteins, J Virol, vol.62, pp.1558-64, 1988.

F. A. Caetano, M. H. Lopes, G. N. Hajj, C. F. Machado, C. Pinto-arantes et al., Endocytosis of prion protein is required for ERK1/2 signaling induced by stress-inducible protein 1, J Neurosci, vol.28, pp.6691-702, 2008.

M. D. Caiati, V. F. Safiulina, G. Fattorini, S. Sivakumaran, G. Legname et al., PrPC controls via protein kinase A the direction of synaptic plasticity in the immature hippocampus, J Neurosci, vol.33, pp.2973-83, 2013.

M. L. Caillet-boudin, F. J. Fernandez-gomez, H. Tran, C. M. Dhaenens, L. Buee et al., Brain pathology in myotonic dystrophy: when tauopathy meets spliceopathy and RNAopathy', Front Mol Neurosci, vol.6, p.57, 2014.

H. A. Cameron and R. D. Mckay, Adult neurogenesis produces a large pool of new granule cells in the dentate gyrus, J Comp Neurol, vol.435, pp.406-423, 2001.

D. Caparros-lefebvre and A. J. Lees, Atypical unclassifiable parkinsonism on Guadeloupe: an environmental toxic hypothesis, Mov Disord, vol.12, pp.114-122, 1920.

A. Cardinale and S. Biocca, Gene-based antibody strategies for prion diseases, Int J Cell Biol, p.710406, 2013.

G. A. Carlson, C. Ebeling, S. L. Yang, G. C. Telling, M. Torchia et al., Prion isolate specified allotypic interactions between the cellular and scrapie prion proteins in congenic and transgenic mice, Proc Natl Acad Sci U S A, vol.91, pp.5690-94, 1994.

B. C. Carlyle, A. C. Nairn, M. Wang, Y. Yang, L. E. Jin et al., cAMP-PKA phosphorylation of tau confers risk for degeneration in aging association cortex, Proc Natl Acad Sci U S A, vol.111, pp.5036-5077, 2014.

R. W. Carrell and D. A. Lomas, Conformational disease, The Lancet, vol.350, pp.134-172, 1997.

J. Castilla, P. Saa, C. Hetz, and C. Soto, In vitro generation of infectious scrapie prions, Cell, vol.121, pp.195-206, 2005.

J. Castilla, P. Saá, R. Morales, K. Abid, K. Maundrell et al., Protein Misfolding Cyclic Amplification for Diagnosis and Prion Propagation Studies, vol.412, pp.3-21, 2006.

B. Caughey and R. E. Race, Potent inhibition of scrapie-associated PrP accumulation by congo red, J Neurochem, vol.59, pp.768-71, 1992.

B. Caughey and G. J. Raymond, Sulfated polyanion inhibition of scrapie-associated PrP accumulation in cultured cells, J Virol, vol.67, pp.643-50, 1993.

B. Caughey, G. J. Raymond, D. Ernst, and R. E. Race, N-terminal truncation of the scrapieassociated form of PrP by lysosomal protease(s): implications regarding the site of conversion of PrP to the protease-resistant state, J Virol, vol.65, pp.6597-603, 1991.

B. Caughey, G. J. Raymond, D. A. Kocisko, and P. T. Lansbury, Scrapie Infectivity Correlates with Converting Activity, Protease Resistance, and Aggregation of Scrapie Associated Prion Protein in Guanidine Denaturation Studies, J Virol, vol.71, pp.4107-4117, 1997.

B. Caughey, L. D. Raymond, G. J. Raymond, L. Maxson, J. Silveira et al., Inhibition of Protease-Resistant Prion Protein Accumulation In Vitro by Curcumin, Journal of Virology, vol.77, pp.5499-502, 2003.

J. Chabry, B. Caughey, and B. Chesebro, Specific inhibition of in vitro formation of proteaseresistant prion protein by synthetic peptides, J Biol Chem, vol.273, pp.13203-13210, 1998.

J. Chabry, C. Ratsimanohatra, I. Sponne, P. P. Elena, J. P. Vincent et al., In Vivo and In Vitro Neurotoxicity of the Human Prion Protein (PrP) Fragment P118 -135 Independently of PrP Expression, J Neurosci, vol.23, pp.462-69, 2003.
URL : https://hal.archives-ouvertes.fr/hal-00091076

M. Chalfie and J. E. Sulston, Developmental Genetics of the Mechanosensory Neurons of Caenorhabditis elegans, Dev Biol, vol.82, pp.358-70, 1981.

M. Chalfie, Y. Tu, G. Euskirchen, W. W. Ward, and D. C. Prasher, Green Fluorescent Protein as a Marker for Gene Expression, Science, vol.263, pp.802-807, 1994.

R. L. Chandler, Encephalopathy in mice produced by inoculation with scrapie brain material, Lancet, vol.1, pp.1378-79, 1961.

F. Checler, Alzheimer's and prion diseases: PDK1 at the crossroads', Nat Med, vol.19, pp.1088-90, 2013.

S. G. Chen and P. Gambetti, A journey through the species barrier, Neuron, vol.34, pp.854-860, 2002.

X. Cheng, Y. Ma, M. Moore, B. A. Hemmings, and S. S. Taylor, Phosphorylation and activation of cAMP-dependent protein kinase by phosphoinositide-dependent protein kinase, Proc Natl Acad Sci U S A, vol.95, pp.9849-54, 1998.

B. Chesebro, R. Race, K. Wehrly, J. Nishio, M. Bloom et al., Identification of scrapie prion protein-specific mRNA in scrapie-infected and uninfected brain, Nature, vol.315, pp.331-364, 1985.

L. B. Chiarini, A. R. Freitas, S. M. Zanata, R. R. Brentani, V. R. Martins et al., Cellular prion protein transduces neuroprotective signals, EMBO J, vol.21, pp.3317-3343, 2002.

S. H. Choi, Y. H. Kim, M. Hebisch, C. Sliwinski, S. Lee et al., A three-dimensional human neural cell culture model of Alzheimer's disease, Nature, vol.515, pp.274-282, 2014.

M. C. Clarke and D. A. Haig, Evidence for the multiplication of scrapie agent in cell culture, Nature, vol.225, pp.100-101, 1970.

N. J. Cobb, F. D. Sonnichsen, H. Mchaourab, and W. K. Surewicz, Molecular architecture of human prion protein amyloid: a parallel, in-register beta-structure', Proc Natl Acad Sci U S A, vol.104, pp.18946-51, 2007.

O. S. Cohen, J. Chapman, A. D. Korczyn, N. Warman-alaluf, Z. Nitsan et al., CSF tau correlates with CJD disease severity and cognitive decline, Acta Neurol Scand, 2015.

A. S. Coitinho, A. R. Freitas, M. H. Lopes, G. N. Hajj, R. Roesler et al., The interaction between prion protein and laminin modulates memory consolidation, Eur J Neurosci, vol.24, pp.3255-64, 2006.

D. W. Colby, Q. Zhang, S. Wang, D. Groth, G. Legname et al., Prion detection by an amyloid seeding assay, Proc Natl Acad Sci U S A, vol.104, pp.20914-20923, 2007.

S. B. Colling, J. Collinge, and J. G. Jefferys, Hippocampal slices from prion protein null mice: disrupted Ca2+-activated K+ currents, Neurosci Letters, vol.209, pp.49-52, 1996.

J. Collinge and A. R. Clarke, A general model of prion strains and their pathogenicity, Science, vol.18, pp.930-966, 2007.

J. Collinge, M. S. Palmer, K. C. Sidle, I. Gowland, R. Medori et al., Transmission of fatal familial insomnia to laboratory animals, Lancet, vol.346, pp.569-70, 1995.

J. Collinge, K. C. Sidle, J. Meads, J. W. Ironside, and A. F. Hill, Molecular analysis of prion strain variation and the aetiology of, p.383, 1996.

J. Collinge, M. A. Whittington, K. C. Sidle, C. J. Smith, M. S. Palmer et al., Prion protein is necessary for normal synaptic function, Nature, vol.370, pp.295-97, 1994.

J. Collinge, M. Gorham, F. Hudson, A. Kennedy, G. Keogh et al., Safety and efficacy of quinacrine in human prion disease (PRION-1 study): a patient-preference trial, The Lancet Neurology, vol.8, pp.334-378, 2009.

S. J. Collins, V. Lewis, M. Brazier, A. F. Hill, A. Fletcher et al., Quinacrine does not prolong survival in a murine Creutzfeldt-Jakob disease model, Ann Neurol, vol.52, pp.503-509, 2002.

P. Cortelli, P. Gambetti, P. Montagna, and E. Lugaresi, Fatal familial insomnia: clinical features and molecular genetics, J Sleep Res, vol.8, pp.23-29, 1999.

C. J. Cortes, K. Qin, J. Cook, A. Solanki, and J. A. Mastrianni, Rapamycin delays disease onset and prevents PrP plaque deposition in a mouse model of Gerstmann-Straussler-Scheinker disease, J Neurosci, vol.32, pp.12396-405, 2012.

M. P. Courageot, N. Daude, R. Nonno, S. Paquet, M. A. Di-bari et al., A cell line infectible by prion strains from different species, J Gen Virol, vol.89, pp.341-348, 2008.

M. Cramm, M. Schmitz, A. Karch, E. Mitrova, F. Kuhn et al., Stability and Reproducibility Underscore Utility of RT-QuIC for Diagnosis of Creutzfeldt-Jakob Disease, Mol Neurobiol, vol.53, pp.1896-904, 2016.

M. Cramm, M. Schmitz, A. Karch, S. Zafar, D. Varges et al., Characteristic CSF prion seeding efficiency in humans with prion diseases, Mol Neurobiol, vol.51, pp.396-405, 2015.

S. Cronier, V. Beringue, A. Bellon, J. M. Peyrin, and H. Laude, Prion strain-and speciesdependent effects of antiprion molecules in primary neuronal cultures, J Virol, vol.81, pp.13794-800, 2007.

S. Cronier, H. Laude, and J. M. Peyrin, Prions can infect primary cultured neurons and astrocytes and promote neuronal cell death, Proc Natl Acad Sci U S A, vol.101, pp.12271-12277, 2004.

C. Crozet, Y. L. Lin, C. Metting, C. Mourton-gilles, P. Corbeau et al., Inhibition of PrPSc formation by lentiviral gene transfer of PrP containing dominant negative mutations, J Cell Sci, vol.117, pp.5591-97, 2004.
URL : https://hal.archives-ouvertes.fr/inserm-00143203

J. Cuillé and P. L. Chelle, La tremblante du mouton est bien inoculable, C. R. Acad. Sci, vol.206, pp.78-79, 1936.

C. D'avanzo, J. Aronson, Y. H. Kim, S. H. Choi, R. E. Tanzi et al., Alzheimer's in 3D culture: challenges and perspectives, Bioessays, vol.37, pp.1139-1187, 2015.

A. S. David, R. Grant, and J. P. Ballantyne, Unsuccessful treatment of Creutzfeldt-Jakob disease with acyclovir, Lancet, vol.1, pp.512-525, 1984.

A. De-luigi, L. Colombo, L. Diomede, R. Capobianco, M. Mangieri et al., The efficacy of tetracyclines in peripheral and intracerebral prion infection, PLoS One, vol.3, p.1888, 2008.

A. De-mario, A. Castellani, C. Peggion, M. L. Massimino, D. Lim et al., The prion protein constitutively controls neuronal store-operated Ca(2+) entry through Fyn kinase, Front Cell Neurosci, vol.9, p.416, 2015.

S. K. Debburman, G. J. Raymond, B. Caughey, and S. Lindquist, Chaperone-supervised conversion of prion protein to its protease-resistant form, Proc Natl Acad Sci U S A, vol.94, pp.13938-13981, 1997.

R. Demaimay, K. T. Adjou, V. Beringue, S. Demart, C. Lasmezas et al., Amphotericin B delays the incubation period of scrapie in intracerebrally inoculated hamsters, J Virol, vol.71, pp.9685-89, 1997.

R. Demaimay, R. Race, and B. Chesebro, Effectiveness of polyene antibiotics in treatment of transmissible spongiform encephalopathy in transgenic mice expressing Syrian hamster PrP only in neurons, J Virol, vol.73, pp.3511-3524, 1999.

P. Deriziotis, R. Andre, D. M. Smith, R. Goold, K. J. Kinghorn et al., Misfolded PrP impairs the UPS by interaction with the 20S proteasome and inhibition of substrate entry, EMBO J, vol.30, pp.3065-77, 2011.

A. Dickinson, Host-pathogen interactions in scrapie, Genetics, vol.79, pp.387-95, 1975.

H. Diringer and B. Ehlers, Chemoprophylaxis of scrapie in mice, J Gen Virol, vol.72, pp.457-60, 1991.

W. M. Dlakic, E. Grigg, and R. A. Bessen, Prion infection of muscle cells in vitro, J Virol, vol.81, pp.4615-4639, 2007.

A. Doble, The pharmacology and mechanism of action of riluzole, Neurology, vol.47, pp.233-274, 1996.

K. Doh-ura, K. Ishikawa, I. Murakami-kubo, K. Sasaki, S. Mohri et al., Treatment of Transmissible Spongiform Encephalopathy by Intraventricular Drug Infusion in Animal Models, Journal of Virology, vol.78, pp.4999-5006, 2004.

K. Doh-ura, T. Iwaki, and B. Caughey, Lysosomotropic agents and cysteine protease inhibitors inhibit scrapie-associated prion protein accumulation, J Virol, vol.74, pp.4894-97, 2000.

D. G. Donne, J. H. Viles, D. Groth, I. Mehlhorn, T. L. James et al., Structure of the recombinant full-length hamster prion protein PrP(29 -231): The N terminus is highly flexible, Proc Natl Acad Sci U S A, vol.94, pp.13452-57, 1997.

C. Duyckaerts, B. Delatour, and M. C. Potier, Classification and basic pathology of Alzheimer disease, Acta Neuropathol, vol.118, pp.5-36, 2009.

J. Edgeworth, M. Ann, A. Farmer, P. Sicilia, J. Tavares et al., Detection of prion infection in variant Creutzfeldt-Jakob disease: a blood-based assay, The Lancet, vol.377, pp.487-93, 2011.

B. Ehlers and H. Diringer, Dextran sulphate 500 delays and prevents mouse scrapie by impairment of agent replication in spleen, J Gen Virol, vol.65, pp.1325-1355, 1984.

M. Enari, E. Flechsig, and C. Weissmann, Scrapie prion protein accumulation by scrapieinfected neuroblastoma cells abrogated by exposure to a prion protein antibody, Proc Natl Acad Sci U S A, vol.98, pp.9295-9304, 2001.

A. Ertmer, S. Gilch, S. W. Yun, E. Flechsig, B. Klebl et al., The tyrosine kinase inhibitor STI571 induces cellular clearance of PrPSc in prion-infected cells, J Biol Chem, vol.279, pp.41918-41945, 2004.

B. A. Faucheux, E. Morain, V. Diouron, J. P. Brandel, D. Salomon et al., Quantification of surviving cerebellar granule neurones and abnormal prion protein (PrPSc) deposition in sporadic Creutzfeldt-Jakob disease supports a pathogenic role for small PrPSc deposits common to the various molecular subtypes, Neuropathol Appl Neurobiol, vol.37, pp.500-512, 2011.

B. A. Faucheux, N. Privat, J. P. Brandel, V. Sazdovitch, J. L. Laplanche et al., Loss of Cerebellar Granule Neurons Is Associated With Punctate but Not With Large Focal Deposits of Prion Proteinnin Creutzfeldt-Jakob Disease, J Neuropathol Exp Neurol, vol.68, pp.892-901, 2009.

D. Fischer, M. D. Mukrasch, J. Biernat, S. Bibow, M. Blackledge et al., Conformational changes specific for pseudophosphorylation at serine 262 selectively impair binding of tau to microtubules, Biochemistry, vol.48, pp.10047-55, 2009.

G. Forloni, N. Angeretti, R. Chiesa, E. Monzani, M. Salmona et al., Neurotoxicity of a prion protein fragment, Nature, vol.362, pp.543-589, 1993.

G. Forloni, S. Iussich, T. Awan, L. Colombo, N. Angeretti et al., Tetracyclines affect prion infectivity, Proc Natl Acad Sci U S A, vol.99, pp.10849-54, 2002.

T. W. Furlow, R. J. Whitley, and F. J. Wilmes, Repeated suppression of Creutzfeldt-Jakob disease with vidarabine, Lancet, vol.2, pp.564-65, 1982.

H. Furukawa, M. Takahashi, M. Nakajima, and M. Yamada, Prospects of the therapeutic approaches to Creutzfeldt-Jakob disease: a clinical trial of antimalarial, vol.60, pp.1649-57, 2002.

D. C. Gajdusek and V. Zigas, Degenerative disease of the central nervous system in New Guinea: the endemic occurrence of "kuru" in the native population, N Engl J Med, vol.257, pp.974-78, 1957.

P. Gambetti, I. Cali, S. Notari, Q. Kong, W. Q. Zou et al., Molecular biology and pathology of prion strains in sporadic human prion diseases, Acta Neuropathol, vol.121, pp.79-90, 2011.

S. Gauczynski, J. M. Peyrin, S. Haïk, C. Leucht, C. Hundt et al., The 37-kDa/67-kDa laminin receptor acts as the cell-surface receptor for the cellular prion protein, EMBO J, vol.20, pp.5863-75, 2001.

B. A. Gavin, M. J. Dolph, N. R. Deleault, J. C. Geoghegan, V. Khurana et al., Accelerated accumulation of misfolded prion protein and spongiform degeneration in a Drosophila model of Gerstmann-Straussler-Scheinker syndrome, J Neurosci, vol.26, pp.12408-12422, 2006.

D. H. Geschwind, A. L. Kuo, K. S. Wong, A. Haman, G. Devereux et al., Quinacrine treatment trial for sporadic Creutzfeldt-Jakob disease, vol.1, pp.2015-2038, 2013.

S. Ghaemmaghami, B. C. May, A. R. Renslo, and S. B. Prusiner, Discovery of 2-aminothiazoles as potent antiprion compounds, J Virol, vol.84, pp.3408-3420, 2010.

B. Ghetti, S. R. Dlouhy, G. Giaccone, O. Bugiani, B. Frangione et al., Gerstmann-Sträussler-Scheinker disease and the Indiana kindred, Brain Pathol, vol.5, pp.61-75, 1995.

B. Ghetti, P. Piccardo, B. Frangione, O. Bugiani, G. Giaccone et al., Prion protein amyloidosis, Brain Pathol, vol.6, pp.127-172, 1996.

B. Ghetti, F. Tagliavini, G. Giaccone, O. Bugiani, B. Frangione et al., Familial Gerstmann-Sträussler-Scheinker disease with neurofibrillary tangles, Mol Neurobiol, vol.8, pp.41-48, 1994.

B. Ghetti, F. Tagliavini, C. L. Masters, K. Beyreuther, G. Giaccone et al., Gerstmann-Sträussler-Scheinker disease. II. Neurofibrillary tangles and plaques with PrP-amyloid coexist in an affected family, Neurology, vol.39, pp.1453-61, 1989.

G. Giaccone, M. Mangieri, R. Capobianco, L. Limido, J. J. Hauw et al., Tauopathy in human and experimental variant Creutzfeldt-Jakob disease, Neurobiol Aging, vol.29, pp.1864-73, 2008.

P. Giannakopoulos, F. R. Herrmann, T. Bussière, C. Bouras, E. Kövari et al.,

J. H. Morrison-perl, C. Gold, and P. R. Hof, Tangle and neuron numbers, but not amyloid load, predict cognitive status in Alzheimer's disease, Neurology, vol.60, pp.1495-500, 2003.

C. J. Gibbs, A. Joy, R. Heffner, M. Franko, M. Miyazaki et al., Clinical and pathological features and laboratory confirmation of Creutzfeldt-Jakob disease in a recipient of pituitary-derived human growth hormone, N Engl J Med, vol.313, pp.734-772, 1985.

A. Giese, M. H. Groschup, B. Hessl, and H. A. Kretzschmar, Neuronal Cell Death in ScrapieInfected Mice Is Due to Apoptosis, Brain Pathol, vol.5, pp.231-252, 1995.

K. Giles, D. B. Berry, C. Condello, B. N. Dugger, Z. Li et al., Optimization of Aryl Amides that Extend Survival in Prion-Infected Mice, J Pharmacol Exp Ther, vol.358, pp.537-584, 2016.

K. Giles, D. B. Berry, C. Condello, R. C. Hawley, A. Gallardo-godoy et al., Different 2-Aminothiazole Therapeutics Produce Distinct Patterns of Scrapie Prion Neuropathology in Mouse Brains, J Pharmacol Exp Ther, vol.355, pp.2-12, 2015.

D. A. Gimbel, H. B. Nygaard, E. E. Coffey, E. C. Gunther, J. Lauren et al., Memory impairment in transgenic Alzheimer mice requires cellular prion protein, J Neurosci, vol.30, pp.6367-74, 2010.

R. K. Giri, R. Young, R. Pitstick, S. J. Dearmond, S. B. Prusiner et al., Prion infection of mouse neurospheres, Proc Natl Acad Sci U S A, vol.103, pp.3875-80, 2006.

M. Goedert, R. Jakes, M. G. Spillantini, M. Hasegawa, M. J. Smith et al., Assembly of microtubule-associated protein tau into Alzheimer-like filaments induced by sulphated glycosaminoglycans, Nature, vol.383, pp.550-553, 1996.
DOI : 10.1038/383550a0

M. C. Golding, C. R. Long, M. A. Carmell, G. J. Hannon, and M. E. Westhusin, Suppression of prion protein in livestock by RNA interference, Proc Natl Acad Sci U S A, vol.103, pp.5285-90, 2006.

W. Goldmann, N. Hunter, G. Benson, J. D. Foster, and J. Hope, Different scrapie-associated fibril proteins (PrP) are encoded by lines of sheep selected for different alleles of the Sip gene, Journal of General Virology, vol.72, pp.2411-2428, 1991.

T. Gómez-isla, R. Hollister, H. West, S. Mui, J. H. Growdon et al., Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer's disease, Ann Neurol, vol.41, pp.17-24, 1997.

C. X. Gong, T. J. Singh, I. Gmndke-iqbal, and K. Iqbal, Phosphoprotein Phosphatase Activities in Alzheimer Disease Brain, J Neurochem, vol.61, pp.921-948, 1993.
DOI : 10.1111/j.1471-4159.1993.tb03603.x

A. Gorkovskiy, K. R. Thurber, R. Tycko, and R. B. Wickner, Locating folds of the in-register parallel beta-sheet of the Sup35p prion domain infectious amyloid, Proc Natl Acad Sci U S A, vol.111, pp.4615-4637, 2014.

C. Govaerts, H. Wille, S. B. Prusiner, and F. E. Cohen, Evidence for assembly of prions with left-handed beta-helices into trimers, Proc Natl Acad Sci U S A, vol.101, pp.8342-8349, 2004.
DOI : 10.1073/pnas.0402254101

URL : http://europepmc.org/articles/pmc420396?pdf=render

E. Graner, A. F. Mercadante, S. M. Zanata, O. V. Forlenza, A. L. Cabral et al., Cellular prion protein binds laminin and mediates neuritogenesis, Mol Brain Res, vol.76, pp.85-92, 2000.
DOI : 10.1016/s0169-328x(99)00334-4

E. Graner, A. F. Mercadante, S. M. Zanata, V. R. Martins, D. G. Jay et al., Laminin-induced PC-12 cell differentiation is inhibited following laser inactivation of cellular prion protein, FEBS Letters, vol.482, pp.257-60, 2000.
DOI : 10.1016/s0014-5793(00)02070-6

URL : https://febs.onlinelibrary.wiley.com/doi/pdf/10.1016/S0014-5793%2800%2902070-6

F. Gray, F. Chrétien, H. Adle-biassette, A. Dorandeu, T. Ereau et al., Neuronal apoptosis in Creutzfeldt-Jakob disease, J Neuropathol Exp Neurol, vol.58, pp.321-349, 1999.

S. M. Greenwood, S. M. Mizielinska, B. G. Frenguelli, J. Harvey, and C. N. Connolly, Mitochondrial dysfunction and dendritic beading during neuronal toxicity, J Biol Chem, vol.282, pp.26235-26279, 2007.
DOI : 10.1074/jbc.m704488200

URL : http://www.jbc.org/content/282/36/26235.full.pdf

J. K. Griffin and N. R. Cashman, Progress in prion vaccines and immunotherapies, Expert Opin Biol Ther, vol.5, pp.97-110, 2005.
DOI : 10.1517/14712598.5.1.97

B. R. Groveman, M. A. Dolan, L. M. Taubner, A. Kraus, R. B. Wickner et al., Parallel in-register intermolecular beta-sheet architectures for prion-seeded prion protein (PrP) amyloids', J Biol Chem, vol.289, pp.24129-24171, 2014.
DOI : 10.1074/jbc.m114.578344

URL : http://www.jbc.org/content/289/35/24129.full.pdf

C. Haass and D. J. Selkoe, Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta-peptide, Nat Rev Mol Cell Biol, vol.8, pp.101-113, 2007.
DOI : 10.1038/nrm2101

S. Haïk and J. P. Brandel, Biochemical and strain properties of CJD prions: complexity versus simplicity, J Neurochem, vol.119, pp.251-61, 2011.

S. Haïk, J. P. Brandel, D. Salomon, V. Sazdovitch, N. Delasnerie-laupretre et al., Compassionate use of quinacrine in Creutzfeldt-Jakob disease fails to show significant effects, Neurology, vol.63, pp.2413-2428, 2004.

S. Haïk, J. M. Peyrin, L. Lins, M. Y. Rosseneu, R. Brasseur et al., Neurotoxicity of the putative transmembrane domain of the prion protein, Neurobiol Dis, vol.7, pp.644-56, 2000.

S. Haïk, G. Marcon, A. Mallet, M. Tettamanti, A. Welaratne et al., Doxycycline in Creutzfeldt-Jakob disease: a phase 2, randomised, doubleblind, vol.13, pp.150-58, 2014.

G. N. Hajj, M. H. Lopes, A. F. Mercadante, S. S. Veiga, R. B. Da-silveira et al., Cellular prion protein interaction with vitronectin supports axonal growth and is compensated by integrins, J Cell Sci, vol.120, pp.1915-1941, 2007.
DOI : 10.1242/jcs.03459

M. Halliday and G. R. Mallucci, Targeting the unfolded protein response in neurodegeneration: A new approach to therapy, Neuropharmacology, pp.76-169, 2014.

D. P. Hanger, B. H. Anderton, and W. Noble, Tau phosphorylation: the therapeutic challenge for neurodegenerative disease', Trends Mol Med, vol.15, pp.112-121, 2009.

S. Hannaoui, A. Gougerot, N. Privat, E. Levavasseur, N. Bizat et al., Cycline efficacy on the propagation of human prions in primary cultured neurons is strain-specific, J Infect Dis, vol.209, pp.1144-1152, 2014.

S. Hannaoui, L. Maatouk, N. Privat, E. Levavasseur, B. A. Faucheux et al., Prion propagation and toxicity occur in vitro with two-phase kinetics specific to strain and neuronal type, J Virol, vol.87, pp.2535-2583, 2013.
DOI : 10.1128/jvi.03082-12

URL : https://jvi.asm.org/content/87/5/2535.full.pdf

T. Haraguchi, S. Fischer, S. Olofsson, T. Endo, D. Groth et al., Asparagine-linked glycosylation of the scrapie and cellular prion proteins, Arch Biochem Biophys, vol.274, pp.1-13, 1989.

H. P. Harding, H. Zeng, Y. Zhang, R. Jungries, P. Chung et al., Diabetes mellitus and exocrine pancreatic dysfunction in perk-/-mice reveals a role for translational control in secretory cell survival, Mol Cell, vol.7, pp.1153-63, 2001.

H. P. Harding, Y. Zhang, A. Bertolotti, H. Zeng, and D. Ron, Perk is essential for translational regulation and cell survival during the unfolded protein response, Mol Cell, vol.5, pp.897-904, 2000.

J. J. Hauw, V. Sazdovitch, J. L. Laplanche, K. Peoc'h, N. Kopp et al., Neuropathologic variants of sporadic Creutzfeldt-Jakob disease and codon 129 of PrP gene, Neurology, vol.54, pp.1641-1687, 2000.

P. A. Hayward, J. E. Bell, and J. W. Ironside, Prion protein immunocytochemistry: reliable protocols for the investigation of Creutzfeldt-Jakob disease, Neuropathol Appl Neurobiol, vol.20, pp.375-83, 1994.

R. S. Hegde, J. A. Mastrianni, M. R. Scott, K. A. Defea, P. Tremblay et al., A Transmembrane Form of the Prion Protein in Neurodegenerative Disease, Science, vol.279, pp.827-861, 1998.

A. Heiseke, Y. Aguib, C. Riemer, M. Baier, and H. M. Schatzl, Lithium induces clearance of protease resistant prion protein in prion-infected cells by induction of autophagy, J Neurochem, vol.109, pp.25-34, 2009.

A. Herbst, P. Banser, C. D. Velasquez, C. E. Mays, V. L. Sim et al., Infectious prions accumulate to high levels in non proliferative C2C12 myotubes, PLoS Pathog, vol.9, p.1003755, 2013.

J. W. Herms, T. Tings, S. Dunker, and H. A. Kretzschmar, Prion protein affects Ca2+-activated K+ currents in cerebellar purkinje cells, Neurobiol Dis, vol.8, pp.324-354, 2001.

J. W. Herms, T. Tings, S. Gall, A. Madlung, A. Giese et al., Evidence of presynaptic location and function of the prion protein, J Neurosci, vol.19, pp.8866-75, 1999.

F. Hernandez, E. Gomez-de-barreda, A. Fuster-matanzo, J. J. Lucas, and J. Avila, GSK3: a possible link between beta amyloid peptide and tau protein, Exp Neurol, vol.223, pp.322-327, 2010.

F. Hernandez, M. Perez, J. J. Lucas, and J. Avila, Sulfo-glycosaminoglycan content affects PHF-tau solubility and allows the identification of different types of PHFs, Brain Res, vol.935, pp.65-72, 2002.

M. E. Herva, A. Relano-gines, A. Villa, and J. M. Torres, Prion infection of differentiated neurospheres, J Neurosci Methods, vol.188, pp.270-275, 2010.

C. Hetz, M. Russelakis-carneiro, K. Maundrell, J. Castilla, and C. Soto, Caspase-12 and endoplasmic reticulum stress mediate neurotoxicity of pathological prion protein, EMBO J, vol.22, pp.5435-5480, 2003.

C. Hetz, K. Maundrell, and C. Soto, Is loss of function of the prion protein the cause of prion disorders?, Trends in Molecular Medicine, vol.9, pp.237-280, 2003.

A. F. Hill, M. Antoniou, and J. Collinge, Protease-resistant prion protein produced in vitro lacks detectable infectivity, J Gen Virol, p.80, 1999.

L. Hong, H. C. Huang, and Z. F. Jiang, Relationship between amyloid-beta and the ubiquitinproteasome system in Alzheimer's disease, Neurol Res, vol.36, pp.276-82, 2014.

M. Hong, V. Zhukareva, V. Vogelsberg-ragaglia, Z. Wszolek, L. Reed et al.,

J. Q. Schellenberg, V. M. Trojanowski, and . Lee, Mutation-Specific Functional Impairments in Distinct Tau Isoforms of Hereditary FTDP-17, Science, vol.282, pp.1914-1931, 1998.

J. J. Hoozemans, E. S. Van-haastert, P. Eikelenboom, R. A. De-vos, J. M. Rozemuller et al., Activation of the unfolded protein response in Parkinson's disease', Biochem Biophys Res Commun, vol.354, pp.707-718, 2007.

J. J. Hoozemans, E. S. Van-haastert, D. A. Nijholt, A. J. Rozemuller, P. Eikelenboom et al., The unfolded protein response is activated in pretangle neurons in Alzheimer's disease hippocampus, Am J Pathol, vol.174, pp.1241-51, 2009.

M. Horiuchi, S. A. Priola, J. Chabry, and B. Caughey, Interactions between heterologous forms of prion protein: binding, inhibition of conversion, and species barriers, Proc Natl Acad Sci U S A, vol.97, pp.5836-5877, 2000.

S. Hornemann, C. Korth, B. Oesch, R. Riek, G. Wider et al., Recombinant full-length murine prion protein,mPrP(23-231): purification and spectroscopic characterization, FEBS Letters, vol.413, pp.277-81, 1997.

E. F. Houston and M. B. Gravenor, Clinical signs in sheep experimentally infected with scrapie and BSE, Vet Rec, vol.152, pp.333-367, 2003.

K. Hsiao, S. R. Dlouhy, M. Farlow, C. Cass, M. Costa et al., Mutant prion proteins in Gerstmann-Sträussler-Scheinker disease with neurofibrillary tangles, Nat Genet, vol.1, pp.68-71, 1992.

K. Hsiao, D. Groth, M. Scott, S. L. Yang, H. Serban et al., Serial transmission in rodents of neurodegeneration from transgenic mice expressing mutant prion protein, Proc Natl Acad Sci U S A, vol.91, pp.9126-9156, 1994.

K. Hsiao, M. Scott, D. Foster, D. Groth, S. J. Dearmond et al., Spontaneous neurodegeneration in transgenic mice with mutant prion protein, Science, vol.250, pp.1587-90, 1990.

Y. Hu, H. Sun, R. T. Owens, Z. Gu, J. Wu et al., Syndecan-1-Dependent Suppression of PDK1/Akt/Bad Signaling by Docosahexaenoic Acid Induces Apoptosis in Prostate Cancer, Neoplasia, vol.12, pp.826-862, 2010.

. Huang, S. B. Ziwei, F. E. Prusiner, and . Cohen, Scrapie prions: a three-dimensional model of an infectious fragment, Folding and Design, vol.1, pp.13-19, 1996.

C. Hundt, J. M. Peyrin, S. Haïk, S. Gauczynski, C. Leucht et al., Identification of interaction domains of the prion protein with its 37-kDa/67-kDa laminin receptor, EMBO J, vol.20, pp.5876-86, 2001.

B. T. Hyman, K. Marzloff, and P. V. Arriagada, The lack of accumulation of senile plaques or amyloid burden in Alzheimer's disease suggests a dynamic balance between amyloid deposition and resolution, J Neuropathol Exp Neurol, vol.52, pp.594-600, 1993.

Y. Ikegaya, J. A. Kim, M. Baba, T. Iwatsubo, N. Nishiyama et al., Rapid and reversible changes in dendrite morphology and synaptic efficacy following NMDA receptor activation: implication for a cellular defense against excitotoxicity, J Cell Sci, vol.114, pp.4083-93, 2001.

M. Ingelsson, H. Fukumoto, K. L. Newell, J. H. Growdon, E. T. Hedley-whyte et al., Early Abeta accumulation and progressive synaptic loss, gliosis, and tangle formation in AD brain, Neurology, vol.62, pp.925-956, 2004.

L. Ingrosso, A. Ladogana, and M. Pocchiari, Congo red prolongs the incubation period in scrapie-infected hamsters, J Virol, vol.69, pp.506-514, 1995.

K. Iqbal, F. Liu, and C. X. Gong, Tau and neurodegenerative disease: the story so far, Nat Rev Neurol, vol.12, pp.15-27, 2016.

J. W. Ironside, D. L. Ritchie, and M. W. Head, Phenotypic variability in human prion diseases, Neuropathol Appl Neurobiol, vol.31, pp.565-79, 2005.

K. Ishizawa, T. Komori, T. Shimazu, T. Yamamoto, T. Kitamoto et al., Hyperphosphorylated tau deposition parallels prion protein burden in a case of GerstmannStraussler-Scheinker syndrome P102L mutation complicated with dementia, Acta Neuropathol, vol.104, pp.342-50, 2002.

S. Itagaki, E. G. Mc-geer, H. Akiyama, S. Zhu, and D. J. Selkoe, Relationship of microglia and astrocytes to amyloid deposits of Alzheimer disease, J Neuroimmunol, vol.24, pp.173-82, 1989.

Y. Iwamaru, T. Takenouchi, K. Ogihara, M. Hoshino, M. Takata et al.,

. Yokoyama, Microglial cell line established from prion protein-overexpressing mice is susceptible to various murine prion strains, J Virol, vol.81, pp.1524-1531, 2007.

G. S. Jackson, I. Murray, L. L. Hosszu, N. Gibbs, J. P. Waltho et al., Location and properties of metal-binding sites on the human prion protein, Proc Natl Acad Sci U S A, vol.98, pp.8531-8536, 2001.

W. S. Jackson, A. W. Borkowski, H. Faas, A. D. Steele, O. D. King et al., Spontaneous generation of prion infectivity in fatal familial insomnia knockin mice, Neuron, vol.63, pp.438-50, 2009.

W. S. Jackson, A. W. Borkowski, N. E. Watson, O. D. King, H. Faas et al., Profoundly different prion diseases in knock-in mice carrying single PrP codon substitutions associated with human diseases, Proc Natl Acad Sci U S A, vol.110, pp.14759-64, 2013.

E. Jaumain, I. Quadrio, L. Herzog, F. Reine, H. Rezaei et al., Absence of Evidence for a Causal Link between Bovine Spongiform Encephalopathy Strain Variant L-BSE and Known Forms of Sporadic Creutzfeldt-Jakob Disease in Human PrP Transgenic Mice, J Virol, vol.90, pp.10867-74, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01602235

M. Jeffrey, I. A. Goodbrand, and C. M. Goodsir, Pathology of the Transmissible Spongiform Encephalopathies with Special Emphasis on Ultrastructure ', Micron, vol.26, pp.277-98, 1995.

J. K. Jeong, M. H. Moon, B. C. Bae, Y. J. Lee, J. W. Seol et al., Autophagy induced by resveratrol prevents human prion protein-mediated neurotoxicity, Neurosci Res, vol.73, pp.99-105, 2012.

D. Jesionek-kupnicka, J. Buczynski, R. Kordek, T. Sobow, I. Kloszewska et al., Programmed cell death (apoptosis) in Alzheimer's disease and CreutzfeldtJakob disease, Folia Neuropathol, vol.35, pp.233-268, 1997.

M. Jin, N. Shepardson, T. Yang, G. Chen, D. Walsh et al., Soluble amyloid betaprotein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration, Proc Natl Acad Sci U S A, vol.108, pp.5819-5843, 2011.
DOI : 10.1073/pnas.1017033108

URL : http://www.pnas.org/content/108/14/5819.full.pdf

D. R. Jones, W. A. Taylor, C. Bate, M. David, and M. Tayebi, A camelid anti-PrP antibody abrogates PrP replication in prion-permissive neuroblastoma cell lines, PLoS One, vol.5, p.9804, 2010.

E. M. Jones and W. K. Surewicz, Fibril conformation as the basis of species-and straindependent seeding specificity of mammalian prion amyloids, Cell, vol.121, pp.63-72, 2005.

Y. O. Kamatari, Y. Hayano, K. Yamaguchi, J. Hosokawa-muto, and K. Kuwata, Characterizing antiprion compounds based on their binding properties to prion proteins: implications as medical chaperones, Protein Sci, vol.22, pp.22-34, 2013.

T. Kampers, P. Friedhoff, J. Biernat, E. M. Mandelkow, and E. Mandelkow, RNA stimulates aggregation of microtubule-associated protein tau into Alzheimer-like paired helical filaments, FEBS Lett, vol.399, pp.344-393, 1996.

E. R. Kandel, The molecular biology of memory: cAMP, PKA, CRE, CREB-1, CREB-2, and CPEB, Mol Brain, p.5, 2012.

K. Kaneko, H. Wille, I. Mehlhorn, H. Zhang, H. Ball et al., Molecular properties of complexes formed between the prion protein and synthetic peptides, J Mol Biol, vol.270, pp.574-86, 1997.

K. Kaneko, L. Zulianello, M. Scott, C. M. Cooper, A. C. Wallace et al., Evidence for protein X binding to a discontinuous epitope on the cellular prion protein during scrapie prion propagation, Proc Natl Acad Sci U S A, vol.94, pp.10069-74, 1997.

Y. E. Karapetyan, G. F. Sferazza, M. Zhou, G. Ottenberg, T. Spicer et al., Unique drug screening approach for prion diseases identifies tacrolimus and astemizole as antiprion agents, Proc Natl Acad Sci U S A, vol.110, pp.7044-7093, 2013.

J. Kazlauskaite, A. Young, C. E. Gardner, J. V. Macpherson, C. Venien-bryan et al., An unusual soluble beta-turn-rich conformation of prion is involved in fibril formation and toxic to neuronal cells, Biochem Biophys Res Commun, vol.328, pp.292-305, 2005.

S. L. Kazmirski, D. Alonso, F. E. Cohen, S. B. Prusiner, and V. Daggett, Theoretical studies of sequence effects on the conformational properties of a fragment of the prion protein: implications for scrapie formation, Chem Biol, vol.2, pp.305-320, 1995.

A. Khalili-shirazi, L. Summers, J. Linehan, G. Mallinson, D. Anstee et al., PrP glycoforms are associated in a strain-specific ratio in native PrPSc, J Gen Virol, vol.86, pp.2635-2679, 2005.

I. Khlistunova, J. Biernat, Y. Wang, M. Pickhardt, M. Bergen et al., Inducible expression of Tau repeat domain in cell models of tauopathy: aggregation is toxic to cells but can be reversed by inhibitor drugs, J Biol Chem, vol.281, pp.1205-1219, 2006.

R. H. Kimberlin and C. Walker, Characteristics of a short incubation model of scrapie in the golden hamster, J Gen Virol, vol.34, pp.295-304, 1977.

H. Kishida, Y. Sakasegawa, K. Watanabe, Y. Yamakawa, M. Nishijima et al., Nonglycosylphosphatidylinositol (GPI)-anchored recombinant prion protein with dominantnegative mutation inhibits PrPScreplicationin vitro, Amyloid, vol.11, pp.14-20, 2004.

T. Kitamoto, J. Tateishi, T. Tashima, I. Takeshita, R. A. Barry et al., Amyloid Plaques in Creutzfeldt-Jakob Disease Stain with Prion Protein Antibodies, Ann Neurol, vol.20, pp.204-212, 1986.

R. Klingenstein, S. Lober, P. Kujala, S. Godsave, S. R. Leliveld et al., Tricyclic antidepressants, quinacrine and a novel, synthetic chimera thereof clear prions by destabilizing detergent-resistant membrane compartments, J Neurochem, vol.98, pp.748-59, 2006.

D. J. Klionsky and S. D. Emr, Autophagy as a regulated pathway of cellular degradation, Science, vol.290, pp.1717-1738, 2000.

P. C. Klohn, L. Stoltze, E. Flechsig, M. Enari, and C. Weissmann, A quantitative, highly sensitive cell-based infectivity assay for mouse scrapie prions, Proc Natl Acad Sci U S A, vol.100, pp.11666-71, 2003.

D. A. Kocisko, J. H. Come, S. A. Priola, B. Chesebro, G. J. Raymond et al., Cell-free formation of protease-resistant prion protein, Nature, vol.370, pp.471-74, 1994.

V. I. Korolchuk, F. M. Menzies, and D. C. Rubinsztein, Mechanisms of cross-talk between the ubiquitin-proteasome and autophagy-lysosome systems, FEBS Lett, vol.584, pp.1393-1401, 2010.

C. Korth, B. C. May, F. E. Cohen, and S. B. Prusiner, Acridine and phenothiazine derivatives as pharmacotherapeutics for prion disease, Proc Natl Acad Sci U S A, vol.98, pp.9836-9877, 2001.

J. I. Kourie, Prion channel proteins and their role in vacuolation and neurodegenerative diseases, Eur Biophys J, vol.31, pp.409-425, 2002.

G. G. Kovacs, M. Puopolo, A. Ladogana, M. Pocchiari, H. Budka et al., Genetic prion disease: the EUROCJD experience, vol.118, pp.166-74, 2005.

G. G. Kovacs, J. Rahimi, T. Strobel, M. I. Lutz, G. Regelsberger et al., Tau Pathology in Creutzfeldt-Jakob Disease Revisited, 2016.

G. G. Kovacs, J. Seguin, I. Quadrio, R. Höftberger, I. Kapas et al., Genetic Creutzfeldt-Jakob disease associated with the E200K mutation: characterization of a complex proteinopathy, Acta Neuropathol, vol.121, pp.39-57, 2011.

J. Kovanen, M. Haltia, and K. Cantell, Failure of interferon to modify Creutzfeldt-Jakob disease', Br Med J, vol.280, p.902, 1980.

B. C. Kraemer, B. Zhang, J. B. Leverenz, J. H. Thomas, J. Q. Trojanowski et al., Neurodegeneration and defective neurotransmission in a Caenorhabditis elegans model of tauopathy, Proc Natl Acad Sci U S A, vol.100, pp.9980-9985, 2003.

H. A. Kretzschmar, S. B. Prusiner, L. E. Stowring, and S. J. Dearmond, Scrapie Prion Proteins Are Synthesized in Neurons, Am J Pathol, vol.122, pp.1-5, 1986.

M. Kristiansen, P. Deriziotis, D. E. Dimcheff, G. S. Jackson, H. Ovaa et al., Disease-associated prion protein oligomers inhibit the 26S proteasome, Mol Cell, vol.26, pp.175-88, 2007.

W. Kudo, H. P. Lee, W. Q. Zou, X. Wang, G. Perry et al., Cellular prion protein is essential for oligomeric amyloid-beta-induced neuronal cell death, Hum Mol Genet, vol.21, pp.1138-1182, 2012.

A. B. Kunnumakkara, D. Bordoloi, G. Padmavathi, J. Monisha, N. K. Roy et al., Curcumin, the golden nutraceutical: multitargeting for multiple chronic diseases, Br J Pharmacol, 2016.

C. Kuwahara, A. M. Takeuchi, T. Nishimura, T. Haraguchi, A. Kubosaki et al., Prions prevent neuronal cell-line death, Nature, vol.400, pp.225-251, 1999.

T. Kuwahara, A. Koyama, K. Gengyo-ando, M. Masuda, H. Kowa et al., Familial Parkinson mutant alpha-synuclein causes dopamine neuron dysfunction in transgenic Caenorhabditis elegans, J Biol Chem, vol.281, pp.334-374, 2006.

A. Ladogana, P. Casaccia, L. Ingrosso, M. Cibati, M. Salvatore et al., Sulphate polyanions prolong the incubation period of scrapie-infected hamsters, J Gen Virol, vol.73, pp.661-65, 1992.

C. Lai, C. Chou, L. Ch'ang, C. Liu, and W. Lin, Identification of Novel Human Genes Evolutionarily Conserved in Caenorhabditis elegans by Comparative Proteomics, Gen Res, vol.10, pp.703-716, 2000.

J. L. Laplanche, N. Delasnerie-laupretre, J. P. Brandel, J. Chatelain, P. Beaudry et al., Molecular genetics of prion diseases in France, Neurology, vol.44, pp.2347-51, 1994.

C. Larramendy-gozalo, A. Barret, E. Daudigeos, E. Mathieu, L. Antonangeli et al., Comparison of CR36, a new heparan mimetic, and pentosan polysulfate in the treatment of prion diseases, J Gen Virol, vol.88, pp.1062-1069, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00109769

E. Laskowska, E. Matuszewska, and D. Kuczy?ska-wi?nik, Small heat shock proteins and protein-misfolding diseases, Curr Pharm Biotechnol, vol.11, pp.146-57, 2010.

C. Lasmezas, J. P. Deslys, R. Demaimay, K. T. Adjou, F. Lamoury et al., BSE transmission to macaques, Nature, vol.381, pp.743-787, 1996.

J. Lauren, D. A. Gimbel, H. B. Nygaard, J. W. Gilbert, and S. M. Strittmatter, Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers, Nature, vol.457, pp.1128-1160, 2009.

H. G. Lee, G. Perry, P. I. Moreira, M. R. Garrett, Q. Liu et al., Tau phosphorylation in Alzheimer's disease: pathogen or protector?', Trends Mol Med, vol.11, pp.164-173, 2005.

J. Li, S. Browning, S. P. Mahal, A. M. Oelschlegel, and C. Weissmann, Darwinian evolution of prions in cell culture, Science, vol.327, pp.869-72, 2010.

S. C. Li, N. K. Goto, K. A. Williams, and C. M. Deber, Alpha-helical, but not beta-sheet, propensity of proline is determined by peptide environment, Proc Natl Acad Sci, pp.6676-81, 1996.

P. P. Liberski, D. C. Gajdusek, and P. Brown, How do neurons degenerate in prion diseases or transmissible spongiform encephalopathies (TSEs): neuronal autophagy revisited, Acta Neurobiol Exp, vol.62, pp.141-188, 2002.

P. P. Liberski, B. Sikorska, J. Bratosiewicz-wasik, D. C. Gajdusek, and P. Brown, Neuronal cell death in transmissible spongiform encephalopathies (prion diseases) revisited: from apoptosis to autophagy, Int J Biochem Cell Biol, vol.36, pp.2473-90, 2004.

D. T. Lin, J. Jodoin, M. Baril, C. G. Goodyer, and A. C. Leblanc, Cytosolic prion protein is the predominant anti-Bax prion protein form: exclusion of transmembrane and secreted prion protein forms in the anti-Bax function, Biochim Biophys Acta, vol.1783, pp.2001-2013, 2008.

C. D. Link, Expression of human beta-amyloid peptide in transgenic Caenorhabditis elegans, Proc Natl Acad Sci U S A, vol.92, pp.9368-72, 1995.

M. H. Lopes, G. N. Hajj, A. G. Muras, G. L. Mancini, R. M. Castro et al., Interaction of cellular prion and stress-inducible protein 1 promotes neuritogenesis and neuroprotection by distinct signaling pathways, J Neurosci, vol.25, pp.11330-11339, 2005.

P. J. Lucassen, A. Williams, W. C. Chung, and H. Fraser, Detection of apoptosis in murine scrapie, Neurosci Lett, vol.198, pp.185-88, 1995.

J. Y. Madec, S. Simon, S. Lezmi, A. Bencsik, J. Grassi et al., Abnormal prion protein in genetically resistant sheep from a scrapie-infected flock, J Gen Virol, vol.85, pp.3483-3489, 2004.

A. C. Magalhaes, G. S. Baron, K. S. Lee, O. Steele-mortimer, D. Dorward et al., Uptake and neuritic transport of scrapie prion protein coincident with infection of neuronal cells, J Neurosci, vol.25, pp.5207-5223, 2005.

E. Magnani, J. Fan, L. Gasparini, M. Golding, M. Williams et al., Interaction of tau protein with the dynactin complex, EMBO J, vol.26, pp.4546-54, 2007.

G. R. Mallucci, S. Ratté, E. A. Asante, J. Linehan, I. Gowland et al., Post-natal knockout of prion protein alters hippocampal CA1 properties, but does not result in neurodegeneration, EMBO J, vol.21, pp.202-212, 2002.

M. Malnar, S. Hecimovic, N. Mattsson, and H. Zetterberg, Bidirectional links between Alzheimer's disease and Niemann-Pick type C disease, Neurobiol Dis, p.72, 2014.

A. Mangé, O. Milhavet, H. E. Mcmahon, D. Casanova, and S. Lehmann, Effect of amphotericin B on wild-type and mutated prion proteins in cultured cells: putative mechanism of action in transmissible spongiform encephalopathies, J Neurochem, vol.74, pp.754-62, 2000.

J. Manson, J. D. West, V. Thomson, P. Mcbride, M. H. Kaufman et al., The prior protein gene: a role in mouse embryogenesis?, Development, vol.115, pp.117-139, 1992.

J. C. Manson, A. R. Clarke, M. L. Hooper, L. Aitchison, I. Mcconnel et al., 129/Ola mice carrying a null mutation in PrP that abolishes mRNA production are developmentally normal, Mol Neurobiol, vol.8, pp.121-148, 1994.

J. C. Manson, J. Hope, A. R. Clarke, A. Johnston, C. Black et al., PrP gene dosage and long term potentiation, Neurodegeneration, vol.4, pp.113-127, 1995.

I. M. Mansuy, Calcineurin in memory and bidirectional plasticity, Biochemical and Biophysical Research Communications, vol.311, pp.1195-208, 2003.

L. Manterola, M. Hernando-rodriguez, A. Ruiz, A. Apraiz, O. Arrizabalaga et al., 1-42 beta-amyloid peptide requires PDK1/nPKC/Rac 1 pathway to induce neuronal death, Transl Psychiatry, vol.3, p.219, 2013.

R. F. Marsh and R. A. Bessen, Epidemiologic and experimental studies on transmissible mink encephalopathy, Dev Biol Stand, vol.80, pp.111-129, 1993.

L. Martin, X. Latypova, and F. Terro, Post-translational modifications of tau protein: implications for Alzheimer's disease, Neurochem Int, vol.58, pp.458-71, 2011.

J. F. Martinez-lage, A. Rabano, J. Bermejo, M. Perez, M. C. Guerrero et al., Creutzfeldt-Jakob disease acquired via a dural graft: failure of therapy with quinacrine and chlorpromazine, Surg Neurol, vol.64, pp.542-547, 2005.

V. R. Martins, R. Linden, M. A. Prado, R. Walz, A. C. Sakamoto et al., Cellular prion protein: on the road for functions, FEBS Letters, vol.512, pp.25-28, 2002.

J. Masel, N. Genoud, and A. Aguzzi, Efficient inhibition of prion replication by PrP-Fc(2) suggests that the prion is a PrP(Sc) oligomer', J Mol Biol, vol.345, pp.1243-51, 2005.

E. Masliah, M. Mallory, L. Hansen, M. Alford, T. Albright et al., Immunoreactivity of CD45, a Protein Phosphotyrosine Phosphatase, Alzheimer's Disease, vol.83, pp.12-20, 1991.

J. A. Mastrianni, The genetics of prion diseases, Genet Med, vol.12, pp.187-95, 2010.

C. Masullo, G. Macchi, Y. G. Xi, and M. Pocchiari, Failure to ameliorate Creutzfeldt-Jakob disease with amphotericin B therapy, J Infect Dis, vol.165, pp.784-85, 1992.

C. K. Mathiason, J. G. Powers, S. J. Dahmes, D. A. Osborn, K. V. Miller et al., Infectious Prions in the Saliva and Blood of Deer with Chronic Wasting Disease, p.314, 2006.

M. P. Mattson, Apoptosis in neurodegenerative disorders, Nat Rev Mol Cell Biol, vol.1, pp.120-149, 2000.

C. E. Mays, C. Kim, T. Haldiman, J. Van-der-merwe, A. Lau et al., Prion disease tempo determined by host-dependent substrate reduction, J Clin Invest, vol.124, pp.847-58, 2014.

L. I. Mcguire, A. H. Peden, C. D. Orru, J. M. Wilham, N. E. Appleford et al., Real time quakinginduced conversion analysis of cerebrospinal fluid in sporadic Creutzfeldt-Jakob disease, Ann Neurol, vol.72, pp.278-85, 2012.

L. I. Mcguire, A. Poleggi, I. Poggliolini, S. Suardi, K. Grznarova et al., Cerebrospinal fluid real-time quaking-induced conversion is a robust and reliable test for sporadic creutzfeldt-jakob disease: An international study, Ann Neurol, vol.80, pp.160-65, 2016.

M. P. Mckinley, B. Hay, V. R. Lingappa, I. Lieberburg, and S. B. Prusiner, Developmental Expression of Prion Protein Gene in Brain, Dev Biol, vol.121, pp.105-115, 1987.

N. F. Mclennan, M. Paul, A. Brennan, I. Mcneill, A. Davies et al., Prion Protein Accumulation and Neuroprotection in Hypoxic Brain Damage, The American Journal of Pathology, vol.165, pp.227-262, 2004.

R. Medori, P. Montagna, H. J. Tritschler, A. Leblanc, P. Cortelli et al., Fatal familial insomnia: a second kindred with mutation of prion protein gene at codon 178, Neurology, vol.42, pp.669-70, 1992.

O. Milhavet, D. Casanova, N. Chevallier, R. D. Mckay, and S. Lehmann, Neural stem cell model for prion propagation, Stem Cells, vol.24, pp.2284-91, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00082370

L. Miller-vedam and S. Ghaemmaghami, Strain specificity and drug resistance in anti-prion therapy, Curr Top Med Chem, vol.13, pp.2397-406, 2013.

G. L. Millhauser, Copper and the prion protein: methods, structures, function, and disease, Annu Rev Phys Chem, vol.58, pp.299-320, 2007.

N. Mitsios, M. Saka, J. Krupinski, R. Pennucci, C. Sanfeliu et al., Cellular prion protein is increased in the plasma and peri-infarcted brain tissue after acute stroke, J Neurosci Res, vol.85, pp.602-613, 2007.

T. Mok, Z. Jaunmuktane, S. Joiner, T. Campbell, C. Morgan et al., Variant CreutzfeldtJakob Disease in a Patient with Heterozygosity at PRNP Codon 129, N Engl J Med, vol.376, pp.292-94, 2017.

P. Montagna, P. Gambetti, P. Cortelli, and E. Lugaresi, Familial and sporadic fatal insomnia, The Lancet Neurology, vol.2, pp.167-76, 2003.
DOI : 10.1016/s1474-4422(03)00323-5

J. A. Moreno, M. Halliday, C. Molloy, H. Radford, N. Verity et al., Oral treatment targeting the unfolded protein response prevents neurodegeneration and clinical disease in prion-infected mice, Sci Transl Med, vol.5, pp.206-138, 2013.

S. Mouillet-richard, M. Ermonval, C. Chebassier, J. L. Laplanche, S. Lehmann et al., Signal transduction through prion protein, Science, vol.289, pp.1925-1963, 2000.
DOI : 10.1126/science.289.5486.1925

S. Mouillet-richard, N. Nishida, E. Pradines, H. Laude, B. Schneider et al., Prions impair bioaminergic functions through serotonin-or catecholamine-derived neurotoxins in neuronal cells, J Biol Chem, vol.283, pp.23782-90, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00331728

S. Mouillet-richard, B. Schneider, E. Pradines, M. Pietri, M. Ermonval et al., Cellular prion protein signaling in serotonergic neuronal cells, Ann N Y Acad Sci, vol.1096, pp.106-125, 2007.

W. E. Müller, H. Ushijima, H. C. Schröder, J. M. Forrest, W. F. Schatton et al., Cytoprotective effect of NMDA receptor antagonists on prion protein (PrionSc)-induced toxicity in rat cortical cell cultures, Eur J Pharmacol, vol.246, pp.261-67, 1993.

M. Nakajima, M. Yamada, T. Kusuhara, H. Furukawa, M. Takahashi et al., Results of quinacrine administration to patients with Creutzfeldt-Jakob disease, Dement Geriatr Cogn Disord, vol.17, pp.158-63, 2004.

N. Naslavsky, . Stein, G. Yanai, A. Friedlander, and . Taraboulos, Characterization of Detergent-insoluble Complexes Containing the Cellular Prion Protein and Its Scrapie Isoform, J Biol Chem, p.272, 1997.

K. E. Nazor, F. Kuhn, T. Seward, M. Green, D. Zwald et al., Immunodetection of disease-associated mutant PrP, which accelerates disease in GSS transgenic mice, EMBO J, vol.24, pp.2472-80, 2005.

G. Neri, L. Figà-talamanca, G. C. Di-battista, and F. Russo, Amantadine in CreutzfeldtJakob disease. Review of the literature and case contribution, Riv Neurobiol, vol.30, pp.47-56, 1984.

J. T. Newington, T. Rappon, S. Albers, D. Y. Wong, R. J. Rylett et al., Overexpression of pyruvate dehydrogenase kinase 1 and lactate dehydrogenase A in nerve cells confers resistance to amyloid beta and other toxins by decreasing mitochondrial respiration and reactive oxygen species production, J Biol Chem, vol.287, pp.37245-58, 2012.

J. T. Nguyen, H. Inouye, M. A. Baldwin, R. J. Fletterick, F. E. Cohen et al., X-ray Diffraction of Scrapie Prion Rods and PrP Peptides, J Mol Biol, vol.252, pp.412-434, 1995.

N. Nishida, D. A. Harris, D. Vilette, H. Laude, Y. Frobert et al., Successful Transmission of Three Mouse-Adapted Scrapie Strains to Murine Neuroblastoma Cell Lines Overexpressing Wild-Type Mouse Prion Protein, J Virol, vol.74, pp.320-345, 2000.

V. Novitskaya, O. V. Bocharova, I. Bronstein, and I. V. Baskakov, Amyloid fibrils of mammalian prion protein are highly toxic to cultured cells and primary neurons, J Biol Chem, vol.281, pp.13828-13864, 2006.

M. Nuvolone, M. Hermann, S. Sorce, G. Russo, C. Tiberi et al., Strictly co-isogenic C57BL/6J-Prnp-/-mice: A rigorous resource for prion science, J Exp Med, vol.213, pp.313-340, 2016.

C. N. O'donovan, D. Tobin, and T. G. Cotter, Prion protein fragment PrP-(106-126) induces apoptosis via mitochondrial disruption in human neuronal SH-SY5Y cells, J Biol Chem, vol.276, pp.43516-43539, 2001.

B. Oesch, D. Westaway, and S. B. Prusiner, Prion protein genes: evolutionary and functional aspects, Curr Top Microbiol Immunol, vol.172, pp.109-133, 1991.

J. M. Oh, H. Y. Shin, S. J. Park, B. H. Kim, J. K. Choi et al., The involvement of cellular prion protein in the autophagy pathway in neuronal cells, Mol Cell Neurosci, vol.39, pp.238-285, 2008.

C. D. Orru, M. Bongianni, G. Tonoli, S. Ferrari, A. G. Hughson et al., A test for Creutzfeldt-Jakob disease using nasal brushings, N Engl J Med, vol.371, pp.519-548, 2014.

C. D. Orru, B. R. Groveman, A. G. Hughson, G. Zanusso, M. B. Coulthart et al., Rapid and sensitive RT-QuIC detection of human Creutzfeldt-Jakob disease using cerebrospinal fluid, p.6, 2015.

C. D. Orru, J. M. Wilham, A. G. Hughson, L. D. Raymond, K. L. Mcnally et al., Human variant Creutzfeldt-Jakob disease and sheep scrapie PrP(res) detection using seeded conversion of recombinant prion protein, Protein Eng Des Sel, vol.22, pp.515-536, 2009.

C. D. Orru, J. M. Wilham, G. J. Raymond, F. Kuhn, B. Schroeder et al., Prion Disease Blood Test Using Immunoprecipitation and Improved Quaking-Induced Conversion, MBio, vol.2, pp.78-89, 2011.

V. G. Ostapchenko, F. H. Beraldo, A. H. Mohammad, Y. F. Xie, P. H. Hirata et al., The prion protein ligand, stress-inducible phosphoprotein 1, regulates amyloid-beta oligomer toxicity, J Neurosci, vol.33, pp.16552-64, 2013.

M. Otto, L. Cepek, P. Ratzka, S. Doehlinger, I. Boekhoff et al., Efficacy of flupirtine on cognitive function in patients with CJD: A double-blind study, Neurology, vol.62, pp.714-732, 2004.

S. Padiolleau-lefevre, C. Alexandrenne, F. Dkhissi, G. Clement, S. Essono et al., Expression and detection strategies for an scFv fragment retaining the same high affinity than Fab and whole antibody: Implications for therapeutic use in prion diseases, Mol Immunol, vol.44, pp.1888-96, 2007.

M. S. Palmer, A. J. Dryden, J. T. Hughes, and J. Collinge, Homozygous prion protein genotype predisposes to sporadic Creutzfeldt-Jakob disease, Nature, p.352, 1991.

B. Pantera, C. Bini, P. Cirri, P. Paoli, G. Camici et al., PrPc activation induces neurite outgrowth and differentiation in PC12 cells: role for caveolin-1 in the signal transduction pathway, J Neurochem, vol.110, pp.194-207, 2009.

P. Parchi, R. Castellani, S. Capellari, B. Ghetti, K. Young et al., Molecular basis of phenotypic variability in sporadic Creutzfeldt-Jakob disease, Ann Neurol, vol.39, pp.767-78, 1996.

P. Parchi, A. Giese, S. Capellari, P. Brown, W. Schulz-schaeffer et al., Classification of Sporadic Creutzfeldt-Jakob Disease Based on Molecular and Phenotypic Analysis of 300 Subjects, Ann Neurol, vol.46, pp.224-257, 1999.

P. Parchi, W. Zou, Y. Wang, P. Brown, S. Capellari et al., Genetic influence on the structural variations of the abnormal prion protein, Proc Natl Acad Sci U S A, vol.97, pp.10168-72, 2000.

A. Parry, I. Baker, R. Stacey, and S. Wimalaratna, Long term survival in a patient with variant Creutzfeldt-Jakob disease treated with intraventricular pentosan polysulphate, J Neurol Neurosurg Psychiatry, vol.78, pp.733-737, 2007.

I. H. Pattison, Experiments with scrapie with special reference to the nature of the agent and the pathology of the disease. In', Slow, Latent and Temperate Virus Infections, NINDB Monograph, vol.2, pp.249-57, 1965.

H. K. Paudel and W. Li, Heparin-induced conformational change in microtubule-associated protein Tau as detected by chemical cross-linking and phosphopeptide mapping, J Biol Chem, vol.274, pp.8029-8067, 1999.

K. Peoc'h, E. Levavasseur, E. Delmont, A. Simone, I. Laffont-proust et al., Substitutions at residue 211 in the prion protein drive a switch between CJD and GSS syndrome, a new mechanism governing inherited neurodegenerative disorders, Hum Mol Genet, vol.21, pp.5417-5445, 2012.

D. Peretz, M. R. Scott, D. Groth, R. A. Williamson, D. R. Burton et al., Strain-specified relative conformational stability of the scrapie prion protein, Protein Sci, vol.10, pp.854-63, 2001.

D. Peretz, R. A. Williamson, K. Kaneko, J. Vergara, E. Leclerc et al., Antibodies inhibit prion propagation and clear cell cultures of prion infectivity, Nature, vol.412, pp.739-782, 2001.

D. Peretz, R. A. Williamson, Y. Matsunaga, H. Serban, C. Pinilla et al., A Conformational Transition at the N Terminus of the Prion Protein Features in Formation of the Scrapie Isoform, J Mol Biol, vol.273, pp.614-636, 1997.

S. Perovic, M. Böhm, E. Meesters, A. Meinhardt, G. Pergande et al., Pharmacological intervention in age-associated brain disorders by Flupirtine: Alzheimer's and prion diseases, Mech Ageing Dev, vol.101, pp.1-19, 1998.

S. Perovic, G. Pergande, H. Ushijima, M. Kelve, J. Forrest et al., Flupirtine partially prevents neuronal injury induced by prion protein fragment and lead acetate, Neurodegeneration, vol.4, pp.369-74, 1995.

S. Perovic, H. C. Schröder, G. Pergande, H. Ushijima, and H. M. Müller, Effect of flupirtine on Bcl-2 and glutathione level in neuronal cells treated in vitro with the prion protein fragment, Exp Neurol, vol.147, pp.518-542, 1997.

V. Perrier, K. Kaneko, J. Safar, J. Vergara, P. Tremblay et al., Dominant-negative inhibition of prion replication in transgenic mice, Proc Natl Acad Sci U S A, vol.99, pp.13079-84, 2002.

V. Perrier, A. C. Wallace, K. Kaneko, J. Safar, S. B. Prusiner et al., Mimicking dominant negative inhibition of prion replication through structure-based drug design, Proc Natl Acad Sci U S A, vol.97, pp.6073-78, 2000.

A. Pfeifer, S. Eigenbrod, S. Al-khadra, A. Hofmann, G. Mitteregger et al., Lentivector-mediated RNAi efficiently suppresses prion protein and prolongs survival of scrapie-infected mice, J Clin Invest, vol.116, pp.3204-3214, 2006.

P. Piccardo, J. Cervenak, O. Yakovleva, L. Gregori, K. Pomeroy et al., Squirrel monkeys (Saimiri sciureus) infected with the agent of bovine spongiform encephalopathy develop tau pathology, J Comp Pathol, vol.147, pp.84-93, 2012.

P. Piccardo, J. C. Manson, D. King, B. Ghetti, and R. M. Barron, Accumulation of prion protein in the brain that is not associated with transmissible disease, Proc Natl Acad Sci U S A, vol.104, pp.4712-4719, 2007.

M. Pietri, C. Dakowski, S. Hannaoui, A. Alleaume-butaux, J. Hernandez-rapp et al., PDK1 decreases TACE-mediated alpha-secretase activity and promotes disease progression in prion and Alzheimer's diseases', Nat Med, vol.19, pp.1124-1155, 2013.

T. Pillot, B. Drouet, M. Pinçon-raymond, J. Vandekerckhove, M. Rosseneu et al., A Nonfibrillar Form of the Fusogenic Prion Protein Fragment [118-135] Induces Apoptotic Cell Death in Rat Cortical Neurons, J Neurochem, vol.75, pp.2298-308, 2000.

E. Planel, K. E. Richter, C. E. Nolan, J. E. Finley, L. Liu et al., Anesthesia leads to tau hyperphosphorylation through inhibition of phosphatase activity by hypothermia, J Neurosci, vol.27, pp.3090-3097, 2007.

M. Pocchiari, S. Schmittinger, and C. Masullo, Amphotericin B delays the incubation period of scrapie in intracerebrally inoculated hamsters, J Gen Virol, vol.68, pp.219-242, 1987.

G. Poli, P. A. Martino, S. Villa, G. Carcassola, M. L. Giannino et al., Evaluation of anti-prion activity of congo red and its derivatives in experimentally infected hamsters, Arzneimittelforschung, vol.54, pp.406-421, 2004.

C. Ponto and I. Zerr, Prionerkrankungen-welche Rolle spielen sie heute?, Info Neurologie & Psychiatrie, vol.15, pp.2-8, 2013.

J. Powell-jackson, R. O. Weller, P. Kennedy, M. A. Preece, E. M. Whitcombe et al., CREUTZFELDT-JAKOB DISEASE AFTER ADMINISTRATION OF HUMAN GROWTH HORMONE, The Lancet, 1985.

E. Pradines, J. Hernandez-rapp, A. Villa-diaz, C. Dakowski, H. Ardila-osorio et al., Pathogenic prions deviate PrP(C) signaling in neuronal cells and impair A-beta clearance, Cell Death Dis, vol.4, p.456, 2013.

E. Pradines, D. Loubet, B. Schneider, J. M. Launay, O. Kellermann et al., CREB-dependent gene regulation by prion protein: impact on MMP-9 and beta-dystroglycan, Cell Signal, vol.20, pp.2050-2058, 2008.

S. B. Prusiner, Shattuck lecture--neurodegenerative diseases and prions, Proc Natl Acad Sci U S A, vol.216, pp.1516-1542, 1982.

S. B. Prusiner, M. Scott, D. Foster, K. M. Pan, D. Groth et al., Transgenetic studies implicate interactions between homologous PrP isoforms in scrapie prion replication, Cell, vol.63, pp.673-86, 1990.

B. E. Race, L. H. Fadness, and B. Chesebro, Characterization of Scrapie Infection in Mouse Neuroblastoma Cells, J Gen Virol, vol.68, pp.1391-99, 1987.

B. Race, K. Phillips, A. Kraus, and B. Chesebro, Phosphorylated human tau associates with mouse prion protein amyloid in scrapie-infected mice but does not increase progression of clinical disease, Prion, vol.10, pp.319-349, 2016.

H. Radford, J. A. Moreno, N. Verity, M. Halliday, and G. R. Mallucci, PERK inhibition prevents tau-mediated neurodegeneration in a mouse model of frontotemporal dementia, Acta Neuropathol, vol.130, pp.633-675, 2015.

A. J. Raeber, T. Muramoto, T. B. Kornberg, and S. B. Prusiner, Expression and targeting of Syrian hamster prion protein induced by heat shock in transgenic Drosophila melanogaster, Mech Dev, vol.51, pp.317-344, 1995.

G. J. Raymond, E. A. Olsen, K. S. Lee, L. D. Raymond, P. K. Bryant et al., Inhibition of proteaseresistant prion protein formation in a transformed deer cell line infected with chronic wasting disease, J Virol, vol.80, pp.596-604, 2006.

L. Reiniger, A. Lukic, J. Linehan, P. Rudge, J. Collinge et al., Tau, prions and A?: the triad of neurodegeneration, Acta Neuropathol, vol.121, pp.5-20, 2011.

A. Relano-gines, S. Lehmann, A. Bencsik, M. E. Herva, J. M. Torres et al., Stem cell therapy extends incubation and survival time in prion-infected mice in a time windowdependant manner, J Infect Dis, vol.204, pp.1038-1083, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00635996

C. H. Reynolds, C. J. Garwood, S. Wray, C. Price, S. Kellie et al., Phosphorylation regulates tau interactions with Src homology 3 domains of phosphatidylinositol 3-kinase, phospholipase Cgamma1, Grb2, and Src family kinases, J Biol Chem, vol.283, pp.18177-86, 2008.

R. Rieger, F. Edenhofer, C. Lasmezas, and S. Weiss, The human 37-kDa laminin receptor precursor interacts with the prion protein in eukaryotic cells, Nat Med, vol.3, pp.1383-88, 1997.

R. Riek, S. Hornemann, G. Wider, M. Billeter, R. Glockshuber et al., NMR structure of the mouse prion protein domain PrP, vol.382, pp.180-82, 1996.

C. Riemer, M. Burwinkel, A. Schwarz, S. Gultner, S. W. Mok et al., Evaluation of drugs for treatment of prion infections of the central nervous system, J Gen Virol, vol.89, pp.594-601, 2008.

M. Roffe, F. H. Beraldo, R. Bester, M. Nunziante, C. Bach et al., Prion protein interaction with stress-inducible protein 1 enhances neuronal protein synthesis via mTOR, Proc Natl Acad Sci U S A, vol.107, pp.13147-52, 2010.

J. Rogers, J. Luber-narod, and S. ,

W. H. Civin-styren, Expression of Immune System-Associated Antigens by Cells of the Human Central Nervous System: Relationship to the Pathology of Alzheimer's Disease, Neurobiol Aging, vol.9, pp.339-388, 1988.

R. Rubenstein, H. Deng, R. E. Race, J. Weina, C. L. Scalli et al., Demonstration of scrapie strain diversity in infected PC12 cells, J Gen Virol, vol.73, pp.3027-3058, 1992.

R. Rubenstein, H. Deng, C. L. Scalli, and M. C. Papini, Alterations in neurotransmitter-related enzyme activity in scrapie-infected PC12 cells, J Gen Virol, vol.72, pp.1279-85, 1991.
DOI : 10.1099/0022-1317-72-6-1279

URL : http://jgv.microbiologyresearch.org/deliver/fulltext/jgv/72/6/JV0720061279.pdf?itemId=/content/journal/jgv/10.1099/0022-1317-72-6-1279&mimeType=pdf&isFastTrackArticle=

P. Saa, J. Castilla, and C. Soto, Ultra-efficient replication of infectious prions by automated protein misfolding cyclic amplification, J Biol Chem, vol.281, pp.35245-52, 2006.

G. P. Saborio, B. Permanne, and C. Soto, Sensitive detection of pathological prion protein by cyclic amplification of protein misfolding, Nature, vol.411, pp.810-823, 2001.

J. Safar, F. E. Cohen, and S. B. Prusiner, Quantitative traits of prion strains are enciphered in the conformation of the prion protein, Arch Virol Suppl, vol.16, pp.227-262, 2000.

J. Safar, H. Wille, V. Itri, D. Groth, H. Serban et al., Eight prion strains have PrPscmolecules with different conformations, Nat Med, vol.4, pp.1157-65, 1998.
DOI : 10.1038/2654

M. K. Sandberg, H. Al-doujaily, B. Sharps, A. R. Clarke, and J. Collinge, Prion propagation and toxicity in vivo occur in two distinct mechanistic phases, Nature, vol.470, pp.540-542, 2011.
DOI : 10.1038/nature09768

M. K. Sandberg, H. Al-doujaily, B. Sharps, M. W. De-oliveira, C. Schmidt et al., Prion neuropathology follows the accumulation of alternate prion protein isoforms after infective titre has peaked, Nat Commun, vol.5, p.4347, 2014.

K. Sano, K. Satoh, R. Atarashi, H. Takashima, Y. Iwasaki et al., Early detection of abnormal prion protein in genetic human prion diseases now possible using real-time QUIC assay, PLoS One, vol.8, p.54915, 2013.

T. G. Santos, I. R. Silva, B. Costa-silva, A. P. Lepique, V. R. Martins et al., Enhanced neural progenitor/stem cells self-renewal via the interaction of stress-inducible protein 1 with the prion protein, Stem Cells, vol.29, pp.1126-1162, 2011.

D. Sarnataro, A. Pepe, G. Altamura, I. Simone, A. Pesapane et al., The 37/67 kDa laminin receptor (LR) inhibitor, NSC47924, affects 37/67 kDa LR cell surface localization and interaction with the cellular prion protein, Sci Rep, vol.6, p.24457, 2016.
DOI : 10.1038/srep24457

URL : https://www.nature.com/articles/srep24457.pdf

H. M. Schatzl, L. Laszlo, D. M. Holtzman, J. Tatzelt, S. J. Dearmond et al., A Hypothalamic Neuronal Cell Line Persistently Infected with Scrapie Prions Exhibits Apoptosis, J Virol, vol.71, pp.8821-8852, 1997.

M. L. Schmidt, V. Zhukareva, K. L. Newell, V. M. Lee, and J. Q. Trojanowski, Tau isoform profile and phosphorylation state in dementia pugilistica recapitulate Alzheimer's disease, Acta Neuropathol, vol.101, pp.518-542, 2001.

A. Schneider, J. Biernat, M. Von-bergen, E. Mandelkow, and E. M. Mandelkow, Phosphorylation that detaches tau protein from microtubules (Ser262, Ser214) also protects it against aggregation into Alzheimer paired helical filaments, Biochemistry, vol.38, pp.3549-58, 1999.
DOI : 10.1021/bi981874p

B. Schneider, V. Mutel, M. Pietri, M. Ermonval, S. Mouillet-richard et al., NADPH oxidase and extracellular regulated kinases 1/2 are targets of prion protein signaling in neuronal and nonneuronal cells, Proc Natl Acad Sci U S A, vol.100, pp.13326-13357, 2003.

S. Schraen-maschke, N. Sergeant, C. M. Dhaenens, S. Bombois, V. Deramecourt et al., Tau as a biomarker of neurodegenerative diseases', vol.2, pp.363-84, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00375314

M. Scott, D. Foster, C. Mirenda, D. Serban, F. Coufal et al., Transgenic mice expressing hamster prion protein produce species-specific scrapie infectivity and amyloid plaques, Cell, vol.59, pp.847-57, 1989.
DOI : 10.1016/0092-8674(89)90608-9

M. R. Scott, J. Safar, G. C. Telling, O. Nguyen, D. Groth et al., Identification of a prion protein epitope modulating transmission of bovine spongiform encephalopathy prions to transgenic mice, Proc Natl Acad Sci U S A, vol.94, pp.14279-84, 1997.

D. Seilhean, C. Duyckaerts, and J. J. Hauw, Fatal familial insomnia and prion diseases, Rev Neurol, vol.151, pp.225-255, 1995.

J. S. Seo, M. H. Moon, J. K. Jeong, J. W. Seol, Y. J. Lee et al., SIRT1, a histone deacetylase, regulates prion protein-induced neuronal cell death, Neurobiol Aging, vol.33, pp.1110-1130, 2012.

J. E. Sepulveda-diaz, S. M. Alavi-naini, M. B. Huynh, M. O. Ouidja, C. Yanicostas et al., HS3ST2 expression is critical for the abnormal phosphorylation of tau in Alzheimer's disease-related tau pathology, Brain, vol.138, pp.1339-54, 2015.

F. Shewmaker, R. B. Wickner, and R. Tycko, Amyloid of the prion domain of Sup35p has an in-register parallel beta-sheet structure, Proc Natl Acad Sci U S A, vol.103, pp.19754-19763, 2006.

W. C. Shyu, S. Z. Lin, M. F. Chiang, D. C. Ding, K. W. Li et al., Overexpression of PrPC by adenovirus-mediated gene targeting reduces ischemic injury in a stroke rat model, J Neurosci, vol.25, pp.8967-77, 2005.

C. J. Sigurdson and M. W. Miller, Other animal prion diseases, Br Med Bull, vol.66, pp.199-212, 2003.

S. Simoneau, H. Rezaei, N. Sales, G. Kaiser-schulz, M. Lefebvre-roque et al., PLoS Pathog, vol.3, p.125, 2007.

T. Skillback, C. Rosen, F. Asztely, N. Mattsson, K. Blennow et al., Diagnostic performance of cerebrospinal fluid total tau and phosphorylated tau in Creutzfeldt-Jakob disease: results from the Swedish Mortality Registry, JAMA Neurol, vol.71, pp.476-83, 2014.

A. D. Snow, R. Kisilevsky, J. Willmer, S. B. Prusiner, and S. J. Dearmond, Sulfated glycosaminoglycans in amyloid plaques of prion diseases, Acta Neuropathol, vol.77, pp.337-379, 1989.

A. D. Snow, T. N. Wight, D. Nochlin, Y. Koike, K. Kimata et al., Immunolocalization of heparan sulfate proteoglycans to the prion protein amyloid plaques of Gerstmann-Straussler syndrome, Creutzfeldt-Jakob disease and scrapie, Lab Invest, vol.63, pp.601-612, 1990.

C. H. Song, H. Furuoka, C. L. Kim, M. Ogino, A. Suzuki et al., Effect of intraventricular infusion of anti-prion protein monoclonal antibodies on disease progression in prion-infected mice, J Gen Virol, vol.89, pp.1533-1577, 2008.

C. Soto, Alzheimer's and prion disease as disorders of protein conformation: implications for the design of novel therapeutic approaches, J Mol Med, vol.77, pp.412-430, 1999.

C. Soto and G. P. Saborio, Prions: disease propagation and disease therapy by conformational transmission', Trends Mol Med, vol.7, pp.109-123, 2001.

C. Soto and N. Satani, The intricate mechanisms of neurodegeneration in prion diseases', Trends Mol Med, vol.17, pp.14-24, 2011.

C. Soto, R. J. Kascsak, G. P. Saborío, P. Aucouturier, T. Wisniewski et al., Reversion of prion protein conformational changes by synthetic b-sheet breaker peptides, The Lancet, vol.355, pp.192-97, 2000.

N. Stahl, M. A. Baldwin, R. Hecker, K. M. Pan, A. L. Burlingame et al., Glycosylinositol phospholipid anchors of the scrapie and cellular prion proteins contain sialic acid, Biochem, vol.31, pp.5043-53, 1992.

J. E. Sulston, E. Schierenberg, J. G. White, and J. N. Thompson, The Embryonic Cell Lineage of the Nematode Caenorhabditis elegans, Dev Biol, vol.100, pp.64-119, 1983.

F. Tagliavini, Prion therapy: Tetracyclic compounds in animal models and patients with Creutzfeldt-Jakob disease, vol.4, pp.149-50, 2008.

F. Tagliavini, G. Forloni, L. Colombo, G. Rossi, L. Girola et al., Tetracycline affects abnormal properties of synthetic PrP peptides and PrP(Sc) in vitro', J Mol Biol, vol.300, pp.1309-1331, 2000.

T. Takenouchi, Y. Iwamaru, M. Sato, T. Yokoyama, and H. Kitani, Establishment of an SV40 large T antigen-immortalized bovine brain cell line and its neuronal differentiation by dibutyryl-cyclic AMP, Cell Biol Int, vol.33, pp.187-91, 2009.

H. Takeuchi, T. Mizuno, G. Zhang, J. Wang, J. Kawanokuchi et al., Neuritic beading induced by activated microglia is an early feature of neuronal dysfunction toward neuronal death by inhibition of mitochondrial respiration and axonal transport, J Biol Chem, vol.280, pp.10444-54, 2005.

G. Tamguney, K. P. Francis, K. Giles, A. Lemus, S. J. Dearmond et al., Measuring prions by bioluminescence imaging, Proc Natl Acad Sci U S A, vol.106, pp.15002-15008, 2009.

A. Taraboulos, A. Serban, and S. B. Prusiner, Scrapie Prion Proteins Accumulate in the Cytoplasm of Persistently Infected Cultured Cells, J Cell Bio, vol.110, pp.2117-2149, 1990.

J. Tateishi, P. Brown, T. Kitamoto, Z. M. Hoque, R. Roos et al., First experimental transmission of fatal familial insomnia, Nature, vol.376, pp.434-469, 1995.

J. Tateishi, T. Kitamoto, M. Z. Hoque, and H. Furukawa, Experimental transmission of Creutzfeldt-Jakob disease and related diseases to rodents, Neurology, vol.46, pp.532-569, 1996.

G. C. Telling, T. Haga, M. Torchia, P. Tremblay, S. J. Dearmond et al., Interactions between wild-type and mutant prion proteins modulate neurodegeneration in transgenic mice, Genes Dev, vol.10, pp.1736-50, 1996.

G. C. Telling, P. Parchi, S. J. Dearmond, P. Cortelli, P. Montagna et al., Evidence for the Conformation of the Pathologic Isoform of the Prion Protein Enciphering and Propagating Prion Diversity, Science, vol.274, pp.2079-82, 1996.

G. C. Telling, M. Scott, K. K. Hsiao, D. Foster, S. L. Yang et al., Transmission of Creutzfeldt-Jakob disease from humans to transgenic mice expressing chimeric human-mouse prion protein, Proc Natl Acad Sci U S A, vol.91, pp.9936-9976, 1994.

G. C. Telling, M. Scott, J. Mastrianni, R. Gabizon, M. Torchia et al., Prion propagation in mice expressing human and chimeric PrP transgenes implicates the interaction of cellular PrP with another protein, Cell, vol.83, pp.79-90, 1995.

T. Terada, Y. Tsuboi, T. Obi, K. Doh-ura, S. Murayama et al., Less protease-resistant PrP in a patient with sporadic CJD treated with intraventricular pentosan polysulphate, Acta Neurol Scand, vol.121, pp.127-157, 2010.

A. M. Thackray, O. Andreoletti, and R. Bujdoso, Bioassay of prion-infected blood plasma in PrP transgenic Drosophila, Biochem J, vol.473, pp.4399-412, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01605089

A. M. Thackray, Y. Di, C. Zhang, H. Wolf, L. Pradl et al., Prion-induced and spontaneous formation of transmissible toxicity in PrP transgenic Drosophila, Biochem J, vol.463, pp.31-40, 2014.

A. M. Thackray, F. Muhammad, C. Zhang, M. Denyer, J. Spiropoulos et al., Prion-induced toxicity in PrP transgenic Drosophila, Exp Mol Pathol, vol.92, pp.194-201, 2012.

A. M. Thackray, F. Muhammad, C. Zhang, Y. Di, T. R. Jahn et al., Ovine PrP transgenic Drosophila show reduced locomotor activity and decreased survival, Biochem J, vol.444, pp.487-95, 2012.

G. Tilly, J. Chapuis, D. Vilette, H. Laude, and J. L. Vilotte, Efficient and specific downregulation of prion protein expression by RNAi, Biochemical and Biophysical Research Communications, vol.305, pp.548-51, 2003.

I. Tobler, T. Deboer, and M. Fischer, Sleep and Sleep Regulation in Normal and Prion ProteinDeficient Mice, J Neurosci, vol.17, pp.1869-79, 1997.

I. Tobler, S. E. Gaus, T. Deboer, P. Achermann, M. Fischer et al., Altered circadian activity rhytms and sleep in mice devoid of prion protein, Nature, vol.380, pp.639-681, 1996.

N. V. Todd, J. Morrow, K. Doh-ura, S. Dealler, S. O'hare et al., Cerebroventricular infusion of pentosan polysulphate in human variant CreutzfeldtJakob disease, J Infect, vol.50, pp.394-400, 2005.

T. Togo, N. Sahara, and S. H. ,

N. Cookson-yen, T. Ishizawa, M. Hutton, R. Silva, A. Lees et al., Argyrophilic grain disease is a sporadic 4-repeat tauopathy, J Neuropathol Exp Neurol, vol.61, pp.547-56, 2002.

M. Toni, E. Spisni, C. Griffoni, S. Santi, M. Riccio et al., Cellular prion protein and caveolin-1 interaction in a neuronal cell line precedes Fyn/Erk 1/2 signal transduction, J Biomed Biotechnol, p.69469, 2006.

J. M. Torres, J. Castilla, B. Pintado, A. Gutierrez-adan, O. Andreoletti et al., Spontaneous generation of infectious prion disease in transgenic mice, Emerg Infect Dis, vol.19, pp.1938-1985, 2013.

K. Toupet, C. V.-compan, C. Crozet, N. Mourton-gilles, F. Mestre-frances et al., Effective gene therapy in a mouse model of prion diseases, PLoS One, vol.3, p.2773, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00322421

C. Tranchant, N. Sergeant, A. Wattez, J. M. Mohr, A. Warter et al., Neurofibrillary tangles in Gerstmann-Sträussler-Scheinker syndrome with the A117V prion gene mutation, J Neurol Neurosurg Psychiatry, vol.63, pp.240-286, 1997.

P. Tremblay, Z. Meiner, M. Galou, C. Heinrich, C. Petromilli et al., Doxycycline control of prion protein transgene expression modulates prion disease in mice, Proc Natl Acad Sci U S A, vol.95, pp.12580-85, 1998.

. Tscs, Genome sequence of the nematode C. elegans: a platform for investigating biology, Science, vol.282, pp.2012-2030, 1998.

Y. Tsuboi, K. Doh-ura, and T. Yamada, Continuous intraventricular infusion of pentosan polysulfate: clinical trial against prion diseases, Neuropathology, vol.29, pp.632-638, 2009.

K. L. Tucker, M. Meyer, and Y. A. Barde, Neurotrophins are required for nerve growth during development, Nat Neurosci, vol.4, pp.29-37, 2001.

E. Turk, D. B. Teplow, L. E. Hood, and S. B. Prusiner, Purification and properties of the cellular and scrapie hamster prion proteins, Eur J Biochem, vol.176, pp.21-30, 1988.

R. Tycko, R. Savtchenko, V. G. Ostapchenko, N. Makarava, and I. V. Baskakov, The alphahelical C-terminal domain of full-length recombinant PrP converts to an in-register parallel beta-sheet structure in PrP fibrils: evidence from solid state nuclear magnetic resonance, Biochemistry, vol.49, pp.9488-97, 2010.

S. Tzaban, G. Friedlander, O. Schonberger, L. Horonchik, Y. Yedidia et al., Protease-Sensitive Scrapie Prion Protein in Aggregates of Heterogeneous Sizes, Biochem, vol.41, pp.12868-75, 2002.

J. W. Um, H. B. Nygaard, J. K. Heiss, M. A. Kostylev, M. Stagi et al., Alzheimer amyloid-beta oligomer bound to postsynaptic prion protein activates Fyn to impair neurons, Nat Neurosci, vol.15, pp.1227-1262, 2012.

E. Uro-coste, H. Cassard, S. Simon, S. Lugan, J. M. Bilheude et al., Beyond PrP9res) type 1/type 2 dichotomy in Creutzfeldt-Jakob disease, PLoS Pathog, vol.4, p.1000029, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00421129

D. L. Vanik, K. A. Surewicz, and W. K. Surewicz, Molecular basis of barriers for interspecies transmissibility of mammalian prions, Mol Cell, vol.14, pp.139-184, 2004.

D. Varges, H. Manthey, U. Heinemann, C. Ponto, M. Schmitz et al., Doxycycline in early CJD: a double-blinded randomised phase II and observational study, J Neurol Neurosurg Psychiatry, 2016.

C. Vidal, C. Herzog, A. M. Haeberle, C. Bombarde, M. C. Miquel et al., Early dysfunction of central 5-HT system in a murine model of bovine spongiform encephalopathy, Neuroscience, vol.160, pp.731-774, 2009.

J. H. Viles, M. Klewpatinond, and R. C. , Copper and the structural biology of the prion protein, Biochem Soc Trans, vol.36, pp.1288-92, 2008.

D. Vilette, Cell models of prion infection, Vet Res, vol.39, p.10, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00902912

D. Vilette, O. Andreoletti, F. Archer, M. F. Madelaine, J. L. Vilotte et al., Ex vivo propagation of infectious sheep scrapie agent in heterologous epithelial cells expressing ovine prion protein, Proc Natl Acad Sci U S A, vol.98, pp.4055-4064, 2001.

J. L. Vilotte, S. Soulier, R. Essalmani, M. G. Stinnakre, D. Vaiman et al., Markedly increased susceptibility to natural sheep scrapie of transgenic mice expressing ovine prp, J Virol, vol.75, pp.5977-84, 2001.

I. Vorberg, A. Raines, B. Story, and S. A. Priola, Susceptibility of Common Fibroblast Cell Lines to Transmissible Spongiform Encephalopathy Agents, J Infect Dis, vol.189, pp.431-470, 2004.

J. D. Wadsworth, E. A. Asante, M. Desbruslais, J. M. Linehan, S. Joiner et al., Human Prion Protein with Valine 129 Prevents Expression of Variant CJD Phenotype, Science, vol.306, pp.1793-96, 2004.

J. Wagner, S. Ryazanov, A. Leonov, J. Levin, S. Shi et al., Anle138b: a novel oligomer modulator for disease-modifying therapy of neurodegenerative diseases such as prion and Parkinson's disease, Acta Neuropathol, vol.125, pp.795-813, 2013.

J. Z. Wang, I. Grundke-iqbal, and K. Iqbal, Glycosylation of microtubule-associated protein tau: an abnormal posttranslational modification in Alzheimer's disease', Nat Med, vol.2, pp.871-75, 1996.

J. Z. Wang and F. Liu, Microtubule-associated protein tau in development, degeneration and protection of neurons, Prog Neurobiol, vol.85, pp.148-75, 2008.

J. Z. Wang and Z. H. Wang, Senescence may mediate conversion of tau phosphorylationinduced apoptotic escape to neurodegeneration, Exp Gerontol, vol.68, pp.82-88, 2015.

J. Wang, B. Y. Zhang, J. Zhang, K. Xiao, L. N. Chen et al., Treatment of SMB-S15 Cells with Resveratrol Efficiently Removes the PrP(Sc) Accumulation In Vitro and Prion Infectivity In Vivo, Mol Neurobiol, vol.53, pp.5367-76, 2016.

X. F. Wang, C. F. Dong, J. Zhang, Y. Z. Wan, F. Li et al., Human tau protein forms complex with PrP and some GSS-and fCJDrelated PrP mutants possess stronger binding activities with tau in vitro, Mol Cell Biochem, vol.310, pp.49-55, 2008.

Y. Wang and E. Mandelkow, Tau in physiology and pathology, Nat Rev Neurosci, vol.17, pp.5-21, 2016.

H. J. Ward, D. Everington, S. N. Cousens, B. Smith-bathgate, M. Leitch et al., Risk factors for variant Creutzfeldt-Jakob disease: a case-control study, Ann Neurol, vol.59, pp.111-131, 2006.

P. Weber, A. Giese, N. Piening, G. Mitteregger, A. Thomzig et al., Cell-free formation of misfolded prion protein with authentic prion infectivity, Proc Natl Acad Sci U S A, vol.103, pp.15818-15841, 2006.

M. D. Weingarten, A. H. Lockwood, S. Hwo, and M. W. Kirschner, A Protein Factor Essential for Microtubule Assembly, Proc Natl Acad Sci U S A, vol.72, pp.1858-62, 1975.

J. Weise, R. O.-crome, W. Sandau, M. Schulz-schaeffer, I. Bahr et al., Upregulation of cellular prion protein (PrPc) after focal cerebral ischemia and influence of lesion severity, Neurosci Lett, vol.372, pp.146-50, 2004.

C. Weissmann, M. Enari, P. C. Klöhn, G. Rossi, and E. Flechsig, Molecular biology of prions, Acta Neurobiol Exp, vol.62, pp.153-66, 2002.

A. R. White, P. Enever, M. Tayebi, R. Mushens, J. Linehan et al., Monoclonal antibodies inhibit prion replication and delay the development of prion disease, Nature, vol.422, pp.80-83, 2003.

J. G. White, E. Southgate, J. N. Thomson, and S. Brenenr, The structure of the nervous system of the nematode Caenorhabditis elegans, Philos Trans R Soc Lond B Biol Sci, vol.314, pp.1-340, 1986.

M. D. White, M. Farmer, I. Mirabile, S. Brandner, J. Collinge et al., Single treatment with RNAi against prion protein rescues early neuronal dysfunction and prolongs survival in mice with prion disease, Proc Natl Acad Sci U S A, vol.105, pp.10238-10281, 2008.

I. R. Whittle, R. S. Knight, and R. G. Will, Unsuccessful intraventricular pentosan polysulphate treatment of variant Creutzfeldt-Jakob disease, Acta Neurochir, vol.148, pp.677-686, 2006.

R. B. Wickner, F. Dyda, and R. Tycko, Amyloid of Rnq1p, the basis of the [PIN+] prion, has a parallel in-register beta-sheet structure, Proc Natl Acad Sci U S A, vol.105, pp.2403-2411, 2008.

J. M. Wilham, C. D. Orru, R. A. Bessen, R. Atarashi, K. Sano et al., Rapid end-point quantitation of prion seeding activity with sensitivity comparable to bioassays, PLoS Pathog, vol.6, p.1001217, 2010.

R. G. Will, J. W. Ironside, M. Zeidler, S. N. Cousens, K. Estibeiro et al., A new variant of Creutzfeldt-Jakob disease in the UK, The Lancet, vol.347, pp.921-946, 1996.

H. Wille, M. D. Michelitsch, V. Guenebaut, S. Supattapone, A. Serban et al., Structural studies of the scrapie prion protein by electron crystallography, Proc Natl Acad Sci U S A, vol.99, pp.3563-3571, 2002.

A. Williams, P. J. Lucassen, D. Ritchie, and M. Bruce, PrP Deposition, Microglial Activation, and Neuronal Apoptosis in Murine Scrapie, Exp Neurol, vol.144, pp.433-471, 1997.

Y. G. Xi, L. Ingrosso, A. Ladogana, C. Masullo, and M. Pocchiari, Amphotericin B treatment dissociates in vivo replication of the scrapie agent from PrP accumulation, Nature, vol.356, pp.598-601, 1992.

X. H. Xu and Z. Zhong, Disease modeling and drug screening for neurological diseases using human induced pluripotent stem cells, Acta Pharmacol Sin, vol.34, pp.755-64, 2013.

R. Zahn, A. Liu, T. Lührs, R. Riek, C. Schroetter et al., NMR solution structure of the human prion protein, Proc Natl Acad Sci U S A, vol.97, pp.145-50, 2000.

S. M. Zanata, M. H. Lopes, A. F. Mercadante, G. N. Hajj, L. B. Chiarini et al., Stress-inducible protein 1 is a cell surface ligand for cellular prion that triggers neuroprotection, EMBO J, vol.21, pp.3307-3323, 2002.

G. Zanusso, M. Fiorini, S. Ferrari, A. Gajofatto, A. Cagnin et al., Cerebrospinal fluid markers in sporadic Creutzfeldt-Jakob disease, Int J Mol Sci, vol.12, pp.6281-92, 2011.

G. Zanusso, S. Monaco, M. Pocchiari, and B. Caughey, Advanced tests for early and accurate diagnosis of Creutzfeldt-Jakob disease, Nat Rev Neurol, vol.12, pp.325-358, 2016.

H. Zhang, K. Kaneko, J. T. Nguyen, T. L. Livshits, M. A. Baldwin et al., Conformational Transitions in Peptides Containing Two Putative alphaHelices of the Prion Protein, J Mol Biol, vol.250, pp.514-540, 1995.

M. Zhou, G. Ottenberg, G. F. Sferrazza, and C. I. Lasmezas, Highly neurotoxic monomeric alpha-helical prion protein, Proc Natl Acad Sci U S A, vol.109, pp.3113-3121, 2012.

C. Zhu, U. S. Herrmann, J. Falsig, I. Abakumova, M. Nuvolone et al., A neuroprotective role for microglia in prion diseases, J Exp Med, vol.213, pp.1047-59, 2016.