T. Jonsson, H. Stefansson, and S. Steinberg, Variant of TREM2 associated with the risk of Alzheimer's disease, N Engl J Med, vol.368, pp.107-116, 2013.

R. Guerreiro, A. Wojtas, and J. Bras, TREM2 variants in Alzheimer's disease, N Engl J Med, vol.368, pp.117-127, 2013.

C. Cruchaga, C. M. Karch, and S. C. Jin, Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer's disease, Nature, vol.505, pp.550-554, 2014.

C. Pottier, D. Hannequin, and S. Coutant, High frequency of potentially pathogenic SORL1 mutations in autosomal dominant early-onset Alzheimer disease, Mol Psychiatry, vol.17, pp.875-879, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00965204

T. Jonsson, J. K. Atwal, and S. Steinberg, A mutation in APP protects against Alzheimer's disease and age-related cognitive decline, Nature, vol.488, pp.96-99, 2012.

M. K. Wetzel-smith, J. Hunkapiller, and T. R. Bhangale, A rare mutation in UNC5C predisposes to late-onset Alzheimer's disease and increases neuronal cell death, Nat Med, vol.20, pp.1452-1457, 2014.

G. Nicolas, C. Charbonnier, and D. Wallon, SORL1 rare variants: a major risk factor for familial early-onset Alzheimer's disease, Mol Psychiatry Epub, 2015.

S. Heilmann, D. Drichel, and J. Clarimon, PLD3 in nonfamilial Alzheimer's disease, Nature, vol.520, pp.3-5, 2015.

B. V. Hooli, C. M. Lill, and K. Mullin, PLD3 gene variants and Alzheimer's disease, Nature, vol.520, pp.7-8, 2015.

J. C. Lambert, B. Grenier-boley, and C. Bellenguez, PLD3 and sporadic Alzheimer's disease risk, Nature, vol.520, p.1, 2015.

S. J. Van-der-lee, H. Holstege, and T. H. Wong, PLD3 variants in population studies, Nature, vol.520, pp.2-3, 2015.

R. Cacace, T. Van-den-bossche, and S. Engelborghs, Rare variants in PLD3 do not affect risk for early-onset Alzheimer disease in a European Consortium Cohort, Hum Mutat, vol.36, pp.1226-1235, 2015.

S. Steinberg, H. Stefansson, and T. Jonsson, Loss-of-function variants in ABCA7 confer risk of Alzheimer's disease, Nat Genet, vol.47, pp.445-447, 2015.

E. Cuyvers, A. De-roeck, and T. Van-den-bossche, ABCA7 in a Belgian cohort of Alzheimer's disease

J. R. Backstrom, G. P. Lim, M. J. Cullen, and Z. A. Tokes, Matrix metalloproteinase-9 (MMP-9) is synthesized in neurons of the human hippocampus and is capable of degrading the amyloid-beta peptide (1-40), J Neurosci, vol.16, pp.7910-7919, 1996.

K. Zou, H. Yamaguchi, H. Akatsu, T. Sakamoto, M. Ko et al., Angiotensin-converting enzyme converts amyloid beta-protein 1-42 (Abeta(142)) to Abeta(1-40), and its inhibition enhances brain Abeta deposition, J Neurosci, vol.27, pp.8-14, 2007.

J. Pacheco-quinto and E. A. Eckman, Endothelin-converting enzymes degrade intracellular beta-amyloid produced within the endosomal/lysosomal pathway and autophagosomes, J Biol Chem, vol.288, pp.5606-5615, 2013.

M. J. Kim, S. S. Chae, Y. H. Koh, S. K. Lee, and J. Sa, Glutamate carboxypeptidase II: an amyloid peptide-degrading enzyme with physiological function in the brain, FASEB J, vol.24, pp.4491-4502, 2010.

S. O. Abdul-hay, T. Sahara, M. Mcbride, D. Kang, and M. A. Leissring, Identification of BACE2 as an avid ss-amyloid-degrading protease, Mol Neurodegener, vol.7, p.46, 2012.

S. Mueller-steiner, Y. Zhou, H. Arai, E. D. Roberson, B. Sun et al., Antiamyloidogenic and neuroprotective functions of cathepsin B: implications for Alzheimer's disease, Neuron, vol.51, pp.703-714, 2006.

C. Y. Lee and G. E. Landreth, The role of microglia in amyloid clearance from the AD brain, J Neural Transm, vol.117, pp.9-13, 2010.

A. Salminen, J. Ojala, A. Kauppinen, K. Kaarniranta, and T. Suuronen, Inflammation in Alzheimer's disease: amyloid-beta oligomers trigger innate immunity defence via pattern recognition receptors, Prog Neurobiol, vol.87, pp.1-8, 2009.

M. T. Heneka, D. T. Golenbock, and E. Latz, Innate immunity in Alzheimer's disease, Nat Immunol, vol.16, issue.2, pp.9-236, 2015.

M. Maier, Y. Peng, L. Jiang, T. J. Seabrook, M. C. Carroll et al., Complement C3 deficiency leads to accelerated amyloid beta plaque deposition and neurodegeneration and modulation of the microglia/macrophage phenotype in amyloid precursor protein transgenic mice, J Neurosci, vol.28, pp.6333-6341, 2008.

H. Crehan, P. Holton, S. Wray, J. Pocock, R. Guerreiro et al., Complement receptor 1 (CR1) and Alzheimer's disease, Immunobiology, vol.217, issue.2, pp.4-250, 2012.

M. T. Heneka, M. J. Carson, E. Khoury, J. Landreth, G. E. Brosseron et al., Neuroinflammation in Alzheimer's disease, Lancet Neurol, vol.14, pp.3-8, 2015.

Y. Wang, M. Cella, K. Mallinson, J. D. Ulrich, K. L. Young et al., TREM2 lipid sensing sustains the microglial response in an Alzheimer's disease model, Cell, vol.160, pp.1061-1071, 2015.

M. Cruts, I. Gijselinck, J. Van-der-zee, S. Engelborghs, H. Wils et al., Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21, Nature, vol.442, pp.9-11, 2006.

M. Baker, I. R. Mackenzie, S. M. Pickering-brown, J. Gass, R. Rademakers et al., Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17, Nature, vol.442, pp.916-919, 2006.

D. C. Perry, M. Lehmann, J. S. Yokoyama, A. Karydas, J. J. Lee et al., Progranulin mutations as risk factors for Alzheimer disease, JAMA Neurol, vol.70, pp.774-778, 2013.

S. S. Minami, S. W. Min, G. Krabbe, C. Wang, Y. Zhou et al., Progranulin protects against amyloid beta deposition and toxicity in Alzheimer's disease mouse models, Nat Med, vol.20, pp.1157-1164, 2014.

S. Steinberg, H. Stefansson, T. Jonsson, H. Johannsdottir, A. Ingason et al., Loss-of-function variants in ABCA7 confer risk of Alzheimer's disease, Nat Genet, vol.47, pp.445-447, 2015.

Q. F. Zhao, J. T. Yu, M. S. Tan, and L. Tan, ABCA7 in Alzheimer's disease, Mol Neurobiol, vol.51, pp.1008-1016, 2015.

K. Satoh, S. Abe-dohmae, S. Yokoyama, G. St, P. Fraser et al., ABCA7 loss of function alters Alzheimer amyloid processing, J Biol Chem, vol.290, pp.24152-24165, 2015.

L. Gregori, C. Fuchs, M. E. Figueiredo-pereira, W. E. Van-nostrand, and D. Goldgaber, Amyloid beta-protein inhibits ubiquitin-dependent protein degradation in vitro, J Biol Chem, vol.270, pp.19702-19708, 1995.

M. P. Burns, L. Zhang, G. W. Rebeck, H. W. Querfurth, and C. E. Moussa, Parkin promotes intracellular Abeta1-42 clearance, Hum Mol Genet, vol.18, pp.3206-3216, 2009.

R. A. Nixon, J. Wegiel, A. Kumar, W. H. Yu, C. Peterhoff et al., Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study, J Neuropathol Exp Neurol, vol.64, pp.1-1, 2005.

F. Pickford, E. Masliah, M. Britschgi, K. Lucin, R. Narasimhan et al., The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice, J Clin Invest, vol.118, pp.2190-2199, 2008.

I. Benilova, E. Karran, D. Strooper, and B. , The toxic Abeta oligomer and Alzheimer's disease: an emperor in need of clothes, Nat Neurosci, vol.15, pp.349-357, 2012.

D. M. Hartley, C. Zhao, A. C. Speier, G. A. Woodard, S. Li et al., Transglutaminase induces protofibril-like amyloid beta-protein assemblies that are proteaseresistant and inhibit long-term potentiation, J Biol Chem, vol.283, pp.16790-16800, 2008.
DOI : 10.1074/jbc.m802215200

URL : http://www.jbc.org/content/283/24/16790.full.pdf

D. Schlenzig, R. Ronicke, H. Cynis, H. H. Ludwig, E. Scheel et al., N-terminal pyroglutamate formation of Abeta38 and Abeta40 enforces oligomer formation and potency to disrupt hippocampal long-term potentiation, J Neurochem, vol.121, pp.774-784, 2012.
DOI : 10.1111/j.1471-4159.2012.07707.x

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1471-4159.2012.07707.x/pdf

S. Kumar, N. Rezaei-ghaleh, D. Terwel, D. R. Thal, M. Richard et al., Extracellular phosphorylation of the amyloid beta-peptide promotes formation of toxic aggregates during the pathogenesis of Alzheimer's disease, EMBO J, vol.30, pp.2255-2265, 2011.

J. Y. Lee, T. B. Cole, R. D. Palmiter, S. W. Suh, and J. Y. Koh, Contribution by synaptic zinc to the gender-disparate plaque formation in human Swedish mutant APP transgenic mice, Proc Natl Acad Sci, vol.99, pp.7705-7710, 2002.

C. G. Evans, S. Wisen, and J. E. Gestwicki, Heat shock proteins 70 and 90 inhibit early stages of amyloid beta-(1-42) aggregation in vitro, J Biol Chem, vol.281, pp.33182-33191, 2006.

J. Ojha, G. Masilamoni, D. Dunlap, R. A. Udoff, and A. G. Cashikar, Sequestration of toxic oligomers by HspB1 as a cytoprotective mechanism, Mol Cell Biol, vol.31, pp.3146-3157, 2011.

M. M. Wilhelmus, W. C. Boelens, I. Otte-holler, B. Kamps, R. M. De-waal et al., Small heat shock proteins inhibit amyloid-beta protein aggregation and cerebrovascular amyloid-beta protein toxicity, Brain Res, vol.1089, pp.6-7, 2006.
DOI : 10.1016/j.brainres.2006.03.058

N. C. Inestrosa, A. Alvarez, C. A. Perez, R. D. Moreno, M. Vicente et al., Acetylcholinesterase accelerates assembly of amyloid-beta-peptides into Alzheimer's fibrils: possible role of the peripheral site of the enzyme, Neuron, vol.16, pp.881-891, 1996.

D. M. Holtzman, K. R. Bales, T. Tenkova, A. M. Fagan, M. Parsadanian et al., Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer's disease, Proc Natl Acad Sci, vol.97, pp.2892-2897, 2000.

S. L. Cotman, W. Halfter, and G. J. Cole, Agrin binds to beta-amyloid (Abeta), accelerates abeta fibril formation, and is localized to Abeta deposits in Alzheimer's disease brain, Mol Cell Neurosci, vol.15, pp.183-198, 2000.
DOI : 10.1006/mcne.1999.0816

G. M. Castillo, C. Ngo, J. Cummings, T. N. Wight, and A. D. Snow, Perlecan binds to the beta-amyloid proteins (A beta) of Alzheimer's disease, accelerates A beta fibril formation, and maintains A beta fibril stability, J Neurochem, vol.69, pp.2452-2465, 1997.

S. Diamant, E. Podoly, A. Friedler, H. Ligumsky, O. Livnah et al., Butyrylcholinesterase attenuates amyloid fibril formation in vitro, Proc Natl Acad Sci, vol.103, pp.8628-8633, 2006.
DOI : 10.1073/pnas.0602922103

URL : http://www.pnas.org/content/103/23/8628.full.pdf

X. Li, X. Zhang, A. R. Ladiwala, D. Du, J. K. Yadav et al., Mechanisms of transthyretin inhibition of beta-amyloid aggregation in vitro, J Neurosci, vol.33, pp.19423-19433, 2013.

T. Nuutinen, T. Suuronen, A. Kauppinen, and A. Salminen, Clusterin: a forgotten player in Alzheimer's disease, Brain Res Rev, vol.61, pp.8-9, 2009.
DOI : 10.1016/j.brainresrev.2009.05.007

S. R. Hughes, O. Khorkova, S. Goyal, J. Knaeblein, J. Heroux et al., Alpha2macroglobulin associates with beta-amyloid peptide and prevents fibril formation, Proc Natl Acad Sci, vol.95, pp.3275-3280, 1998.
DOI : 10.1073/pnas.95.6.3275

URL : http://www.pnas.org/content/95/6/3275.full.pdf

S. A. Kaeser, M. C. Herzig, J. Coomaraswamy, E. Kilger, M. L. Selenica et al., Cystatin C modulates cerebral beta-amyloidosis, Nat Genet, vol.39, pp.1437-1439, 2007.
DOI : 10.1038/ng.2007.23

W. Mi, M. Pawlik, M. Sastre, S. S. Jung, D. S. Radvinsky et al., Cystatin C inhibits amyloid-beta deposition in Alzheimer's disease mouse models, Nat Genet, vol.39, pp.1440-1442, 2007.
DOI : 10.1038/ng.2007.29

S. Eriksson, S. Janciauskiene, and L. Lannfelt, Alpha 1-antichymotrypsin regulates Alzheimer beta-amyloid peptide fibril formation, Proc Natl Acad Sci, vol.92, pp.2313-2317, 1995.
DOI : 10.1073/pnas.92.6.2313

URL : http://www.pnas.org/content/92/6/2313.full.pdf

S. Kocherhans, A. Madhusudan, J. Doehner, K. S. Breu, R. M. Nitsch et al., Reduced reelin expression accelerates amyloid-beta plaque formation and tau pathology in transgenic Alzheimer's disease mice, J Neurosci, vol.30, pp.9228-9240, 2010.
DOI : 10.1523/jneurosci.0418-10.2010

URL : http://www.jneurosci.org/content/30/27/9228.full.pdf

M. Jucker and L. C. Walker, Pathogenic protein seeding in Alzheimer disease and other neurodegenerative disorders, Ann Neurol, vol.70, pp.532-540, 2011.
DOI : 10.1002/ana.22615

URL : http://europepmc.org/articles/pmc3203752?pdf=render

E. Masliah, E. Rockenstein, I. Veinbergs, Y. Sagara, M. Mallory et al., Beta-amyloid peptides enhance alpha-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer's disease and Parkinson's disease, Proc Natl Acad Sci, vol.98, pp.12245-12250, 2001.

K. Ono, R. Takahashi, T. Ikeda, and M. Yamada, Cross-seeding effects of amyloid betaprotein and alpha-synuclein, J Neurochem, vol.122, issue.8, pp.3-890, 2012.
DOI : 10.1016/j.jalz.2014.05.1152

URL : https://kanazawa-u.repo.nii.ac.jp/?action=repository_action_common_download&item_id=13927&item_no=1&attribute_id=26&file_no=1

Y. S. Fang, K. J. Tsai, Y. J. Chang, P. Kao, R. Woods et al., Full-length TDP-43 forms toxic amyloid oligomers that are present in frontotemporal lobar dementia-TDP patients, Nat Commun, vol.5, p.4824, 2014.
DOI : 10.1038/ncomms5824

URL : http://www.nature.com/articles/ncomms5824.pdf

M. J. Guerrero-munoz, D. L. Castillo-carranza, S. Krishnamurthy, A. A. Paulucci-holthauzen, U. Sengupta et al., Amyloid-beta oligomers as a template for secondary amyloidosis in Alzheimer's disease, Neurobiol Dis, vol.71, pp.14-23, 2014.

K. A. Josephs, J. L. Whitwell, S. D. Weigand, M. E. Murray, N. Tosakulwong et al., TDP-43 is a key player in the clinical features associated with Alzheimer's disease, Acta Neuropathol, vol.127, pp.811-824, 2014.

, Modeling an A?-centered biological network D Campion et al

R. L. Hamilton, Lewy bodies in Alzheimer's disease: a neuropathological review of 145 cases using alpha-synuclein immunohistochemistry, Brain Pathol, vol.10, pp.378-384, 2000.

M. Rapoport, H. N. Dawson, L. I. Binder, M. P. Vitek, and A. Ferreira, Tau is essential to beta-amyloid-induced neurotoxicity, Proc Natl Acad Sci, vol.99, pp.6364-6369, 2002.

E. D. Roberson, K. Scearce-levie, J. J. Palop, F. Yan, I. H. Cheng et al., Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer's disease mouse model, Science, vol.316, issue.7, pp.0-754, 2007.

L. M. Ittner, Y. D. Ke, F. Delerue, M. Bi, A. Gladbach et al., Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer's disease mouse models, Cell, vol.142, pp.387-397, 2010.

M. W. Salter and L. V. Kalia, Src kinases: a hub for NMDA receptor regulation, Nat Rev Neurosci, vol.5, pp.317-328, 2004.

R. M. Nisbet, J. C. Polanco, L. M. Ittner, and J. Gotz, Tau aggregation and its interplay with amyloid-beta, Acta Neuropathol, vol.129, pp.207-220, 2015.

S. Tu, S. Okamoto, S. A. Lipton, and H. Xu, Oligomeric Abeta-induced synaptic dysfunction in Alzheimer's disease, Mol Neurodegener, vol.9, pp.4-8, 2014.

T. L. Spires-jones, T. Friedman, R. Pitstick, M. Polydoro, A. Roe et al., Methylene blue does not reverse existing neurofibrillary tangle pathology in the rTg4510 mouse model of tauopathy, Neurosci Lett, vol.562, pp.6-9, 2014.

M. G. Spillantini and M. Goedert, Tau pathology and neurodegeneration, Lancet Neurol, vol.12, pp.609-622, 2013.

H. Zempel and E. Mandelkow, Lost after translation: missorting of tau protein and consequences for Alzheimer disease, Trends Neurosci, vol.37, pp.7-9, 2014.

M. L. Frandemiche, D. Seranno, S. Rush, T. Borel, E. Elie et al., Activitydependent tau protein translocation to excitatory synapse is disrupted by exposure to amyloid-beta oligomers, J Neurosci, vol.34, pp.6084-6097, 2014.

W. Noble, D. P. Hanger, C. C. Miller, and S. Lovestone, The importance of tau phosphorylation for neurodegenerative diseases, Front Neurol, vol.4, p.3, 2013.

K. Iqbal, C. X. Gong, and F. Liu, Hyperphosphorylation-induced tau oligomers, Front Neurol, vol.4, p.112, 2013.

S. Kins, A. Crameri, D. R. Evans, B. A. Hemmings, R. M. Nitsch et al., Reduced protein phosphatase 2 A activity induces hyperphosphorylation and altered compartmentalization of tau in transgenic mice, J Biol Chem, vol.276, pp.38193-38200, 2001.

W. Yu, J. Polepalli, D. Wagh, J. Rajadas, R. Malenka et al., A critical role for the PAR-1/ MARK-tau axis in mediating the toxic effects of Abeta on synapses and dendritic spines, Hum Mol Genet, vol.21, pp.1-3, 2012.

G. Mairet-coello, J. Courchet, S. Pieraut, V. Courchet, A. Maximov et al., The CAMKK2-AMPK kinase pathway mediates the synaptotoxic effects of Abeta oligomers through Tau phosphorylation, Neuron, vol.78, pp.9-13, 2013.

M. S. Lee, Y. T. Kwon, M. Li, J. Peng, R. M. Friedlander et al., Neurotoxicity induces cleavage of p35 to p25 by calpain, Nature, vol.405, pp.3-6, 2000.

J. A. Hartigan and G. V. Johnson, Transient increases in intracellular calcium result in prolonged site-selective increases in Tau phosphorylation through a glycogen synthase kinase 3beta-dependent pathway, J Biol Chem, vol.274, pp.21395-21401, 1999.

R. Kimura, K. Kamino, M. Yamamoto, A. Nuripa, T. Kida et al., The DYRK1A gene, encoded in chromosome 21 Down syndrome critical region, bridges between beta-amyloid production and tau phosphorylation in Alzheimer disease, Hum Mol Genet, vol.16, pp.1-5, 2007.

D. Del-prete, F. Checler, and M. Chami, Ryanodine receptors: physiological function and deregulation in Alzheimer disease, Mol Neurodegen, vol.9, issue.2 1, 2014.

E. Ferreiro, C. R. Oliveira, and C. Pereira, Involvement of endoplasmic reticulum Ca 2+ release through ryanodine and inositol 1,4,5-triphosphate receptors in the neurotoxic effects induced by the amyloid-beta peptide, J Neurosci Res, vol.76, pp.872-880, 2004.

U. Dreses-werringloer, J. C. Lambert, V. Vingtdeux, H. Zhao, H. Vais et al., A polymorphism in CALHM1 influences Ca2+ homeostasis, Abeta levels, and Alzheimer's disease risk, Cell, vol.133, pp.1149-1161, 2008.

T. C. Gamblin, F. Chen, A. Zambrano, A. Abraha, S. Lagalwar et al., Caspase cleavage of tau: linking amyloid and neurofibrillary tangles in Alzheimer's disease, Proc Natl Acad Sci, vol.100, pp.10032-10037, 2003.

J. L. Guo and V. M. Lee, Seeding of normal tau by pathological tau conformers drives pathogenesis of Alzheimer-like tangles, J Biol Chem, vol.286, pp.15317-15331, 2011.

J. P. Guo, T. Arai, J. Miklossy, and P. L. Mcgeer, Abeta and tau form soluble complexes that may promote self aggregation of both into the insoluble forms observed in Alzheimer's disease, Proc Natl Acad Sci, vol.103, pp.1953-1958, 2006.

H. Hsieh, J. Boehm, C. Sato, T. Iwatsubo, T. Tomita et al., AMPAR removal underlies Abeta-induced synaptic depression and dendritic spine loss, Neuron, vol.52, pp.831-843, 2006.

H. Y. Wang, D. H. Lee, D. Andrea, M. R. Peterson, P. A. Shank et al., Beta-amyloid (1-42) binds to alpha7 nicotinic acetylcholine receptor with high affinity. Implications for Alzheimer's disease pathology, J Biol Chem, vol.275, pp.5-6, 2000.

M. Larson, M. A. Sherman, F. Amar, M. Nuvolone, J. A. Schneider et al., The complex PrP(c)-Fyn couples human oligomeric Abeta with pathological tau changes in Alzheimer's disease, J Neurosci, vol.32, pp.16857-71, 2012.

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.1227-1235, 2012.

T. Kim, G. S. Vidal, M. Djurisic, C. M. William, M. E. Birnbaum et al., Human LilrB2 is a beta-amyloid receptor and its murine homolog PirB regulates synaptic plasticity in an Alzheimer's model, Science, vol.341, pp.1399-1404, 2013.

J. K. Knowles, J. Rajadas, T. V. Nguyen, T. Yang, M. C. Lemieux et al., The p75 neurotrophin receptor promotes amyloid-beta(1-42)-induced neuritic dystrophy in vitro and in vivo, J Neurosci, vol.29, pp.10627-10637, 2009.

M. Cisse and F. Checler, Eph receptors: new players in Alzheimer's disease pathogenesis, Neurobiol Dis, vol.73, pp.1-3, 2015.

J. W. Um, A. C. Kaufman, M. Kostylev, J. K. Heiss, M. Stagi et al., Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer abeta oligomer bound to cellular prion protein, Neuron, vol.79, issue.8, pp.7-902, 2013.

F. Roselli, M. Tirard, J. Lu, P. Hutzler, P. Lamberti et al., Soluble betaamyloid1-40 induces NMDA-dependent degradation of postsynaptic density-95 at glutamatergic synapses, J Neurosci, vol.25, pp.11061-11070, 2005.

G. M. Shankar, B. L. Bloodgood, M. Townsend, D. M. Walsh, D. J. Selkoe et al., Natural oligomers of the Alzheimer amyloid-beta protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway, J Neurosci, vol.27, pp.2866-2875, 2007.

G. M. Mckhann, D. S. Knopman, H. Chertkow, B. T. Hyman, C. R. Jack et al., The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease, Alzheimers Dement J Alzheimers Assoc, vol.7, pp.263-269, 2011.

R. Ossenkoppele, W. J. Jansen, G. D. Rabinovici, D. L. Knol, W. M. Van-der-flier et al., Prevalence of amyloid PET positivity in dementia syndromes: a meta-analysis, JAMA, vol.313, pp.1939-1949, 2015.

C. R. Jack, D. S. Knopman, G. Chételat, D. Dickson, A. M. Fagan et al., Suspected non-Alzheimer disease pathophysiology-concept and controversy, Nat Rev Neurol, vol.12, pp.1-1, 2016.

C. R. Jack, D. S. Knopman, S. D. Weigand, H. J. Wiste, P. Vemuri et al., An operational approach to National Institute on Aging-Alzheimer's Association criteria for preclinical Alzheimer disease, Ann Neurol, vol.71, pp.7-13, 2012.

E. Cuyvers, A. De-roeck, T. Van-den-bossche, C. Van-cauwenberghe, K. Bettens et al., Mutations in ABCA7 in a Belgian cohort of Alzheimer's disease patients: a targeted resequencing study, Lancet Neurol, vol.14, pp.814-822, 2015.

R. Guerreiro, A. Wojtas, J. Bras, M. Carrasquillo, E. Rogaeva et al., TREM2 variants in Alzheimer's disease, N Engl J Med, vol.368, pp.1-1, 2013.

T. Jonsson, H. Stefansson, S. Steinberg, I. Jonsdottir, P. V. Jonsson et al., Variant of TREM2 associated with the risk of Alzheimer's disease, N Engl J Med, vol.368, pp.1-1, 2013.

L. Guennec, K. , N. G. Quenez, O. Charbonnier, C. Wallon et al., ABCA7 rare variants and Alzheimer disease risk, Neurology, vol.86, pp.2134-2137, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01831744

G. Nicolas, C. Charbonnier, D. Wallon, O. Quenez, C. Bellenguez et al., SORL1 rare variants: a major risk factor for familial early-onset Alzheimer's disease, Mol Psychiatry, vol.21, pp.3-4, 2016.

S. Steinberg, H. Stefansson, T. Jonsson, H. Johannsdottir, A. Ingason et al., Loss-of-function variants in ABCA7 confer risk of Alzheimer's disease, Nat Genet, vol.47, pp.445-447, 2015.

M. Hiltunen, S. Helisalmi, A. Mannermaa, I. Alafuzoff, A. M. Koivisto et al., Identification of a novel 4.6-kb genomic deletion in presenilin-1 gene which results in exclusion of exon 9 in a Finnish early onset Alzheimer's disease family: an Alu core sequence-stimulated recombination?, Eur J Hum Genet EJHG, vol.8, pp.259-266, 2000.

G. Prihar, A. Verkkoniem, J. Perez-tur, R. Crook, S. Lincoln et al., Alzheimer disease PS-1 exon 9 deletion defined, Nat Med, vol.5, p.1090, 1999.

A. Rovelet-lecrux, D. Hannequin, G. Raux, L. Meur, N. Laquerrière et al., APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy, Nat Genet, vol.38, pp.2-4, 2006.

B. V. Hooli, Z. M. Kovacs-vajna, K. Mullin, M. A. Blumenthal, M. Mattheisen et al., Rare autosomal copy number variations in early-onset familial Alzheimer's disease, Mol Psychiatry, vol.19, pp.6-7, 2014.

A. Rovelet-lecrux, S. Legallic, D. Wallon, J. Flaman, O. Martinaud et al., A genome-wide study reveals rare CNVs exclusive to extreme phenotypes of Alzheimer disease, Eur J Hum Genet EJHG, vol.20, pp.613-617, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00965208

N. Brouwers, C. Van-cauwenberghe, S. Engelborghs, J. Lambert, K. Bettens et al., Alzheimer risk associated with a copy number variation in the complement receptor 1 increasing C3b/C4b binding sites, Mol Psychiatry, vol.17, pp.223-233, 2012.

G. Nicolas, D. Wallon, C. Charbonnier, O. Quenez, S. Rousseau et al., Screening of dementia genes by whole-exome sequencing in early-onset Alzheimer disease: input and lessons, Eur J Hum Genet EJHG, vol.24, pp.7-8, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01431285

M. Dejesus-hernandez, I. R. Mackenzie, B. F. Boeve, A. L. Boxer, M. Baker et al., Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS, Neuron, vol.72, issue.2, pp.5-256, 2011.

A. Mckenna, M. Hanna, E. Banks, A. Sivachenko, K. Cibulskis et al., The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data, Genome Res, vol.20, pp.1297-1303, 2010.

H. Li and R. Durbin, Fast and accurate long-read alignment with Burrows-Wheeler transform, Bioinforma Oxf Engl, vol.26, pp.5-8, 2010.

C. A. Anderson, F. H. Pettersson, G. M. Clarke, L. R. Cardon, A. P. Morris et al., Data quality control in genetic case-control association studies, Nat Protoc, 2010.

C. C. Chang, C. C. Chow, L. C. Tellier, S. Vattikuti, S. M. Purcell et al., Second-generation PLINK: rising to the challenge of larger and richer datasets, GigaScience, vol.4, p.7, 2015.

G. Jun, M. Flickinger, K. N. Hetrick, J. M. Romm, K. F. Doheny et al., Detecting and estimating contamination of human DNA samples in sequencing and array-based genotype data, Am J Hum Genet, vol.91, pp.3-9, 2012.

D. Backenroth, J. Homsy, L. R. Murillo, J. Glessner, E. Lin et al., CANOES: detecting rare copy number variants from whole exome sequencing data, Nucleic Acids Res, vol.42, p.97, 2014.

S. Swaminathan, M. J. Huentelman, J. J. Corneveaux, A. J. Myers, K. M. Faber et al., Analysis of copy number variation in Alzheimer's disease in a cohort of clinically characterized and neuropathologically verified individuals, PloS One, vol.7, p.50640, 2012.

S. Swaminathan, L. Shen, S. Kim, M. Inlow, J. D. West et al., Analysis of copy number variation in Alzheimer's disease: the NIALOAD/ NCRAD Family Study, Curr Alzheimer Res, vol.9, pp.801-814, 2012.

J. Alexander, O. Kalev, S. Mehrabian, L. Traykov, M. Raycheva et al., Familial early-onset dementia with complex neuropathologic phenotype and genomic background, Neurobiol Aging, vol.42, pp.1-9, 2016.

A. Rovelet-lecrux, D. Hannequin, O. Guillin, S. Legallic, S. Jurici et al., Frontotemporal dementia phenotype associated with MAPT gene duplication, J Alzheimers Dis JAD, vol.21, pp.8-9, 2010.

D. Campion, C. Pottier, N. G. , L. Guennec, K. Rovelet-lecrux et al., Alzheimer disease: modeling an A?-centered biological network, Mol Psychiatry, vol.21, pp.861-871, 2016.

B. P. Coe, K. Witherspoon, J. A. Rosenfeld, B. Van-bon, A. T. Vulto-van-silfhout et al., Refining analyses of copy number variation identifies specific genes associated with developmental delay, Nat Genet, vol.46, pp.1063-1071, 2014.

D. M. Ruderfer, T. Hamamsy, M. Lek, K. J. Karczewski, D. Kavanagh et al., Patterns of genic intolerance of rare copy number variation in 59,898 human exomes, Nat Genet, vol.48, issue.1, pp.0-7, 2016.

C. R. Jack and D. M. Holtzman, Biomarker modeling of Alzheimer's disease, Neuron, vol.80, pp.1347-1358, 2013.

F. H. Duits, C. E. Teunissen, F. H. Bouwman, P. Visser, N. Mattsson et al., The cerebrospinal fluid 'Alzheimer profile': easily said, but what does it mean?, Alzheimers Dement J Alzheimers Assoc, vol.10, pp.7-8, 2014.

M. G. Spillantini and M. Goedert, Tau pathology and neurodegeneration, Lancet Neurol, vol.12, pp.0-9, 2013.
DOI : 10.1016/s1474-4422(13)70090-5

M. Allen, M. Kachadoorian, Z. Quicksall, F. Zou, H. S. Chai et al., Association of MAPT haplotypes with Alzheimer's disease risk and MAPT brain gene expression levels, Alzheimers Res Ther, vol.6, p.39, 2014.

F. Natacci, E. Alfei, L. Tararà, D. 'arrigo, S. Zuffardi et al., Chromosome 17q21.31 duplication syndrome: Description of a new familiar case and further delineation of the clinical spectrum, Eur J Paediatr Neurol EJPN Off J Eur Paediatr Neurol Soc, vol.20, pp.1-8, 2016.

S. Kitsiou-tzeli, H. Frysira, K. Giannikou, A. Syrmou, K. Kosma et al., Microdeletion and microduplication 17q21.31 plus an additional CNV, in patients with intellectual disability, identified by array-CGH, Gene, vol.492, pp.3-4, 2012.

M. Kirchhoff, A. Bisgaard, M. Duno, F. J. Hansen, and M. Schwartz, A 17q21.31 microduplication, reciprocal to the newly described 17q21.31 microdeletion, in a girl with severe psychomotor developmental delay and dysmorphic craniofacial features, Eur J Med Genet, vol.50, issue.2, pp.6-263, 2007.

B. Grisart, L. Willatt, A. Destrée, J. Fryns, K. Rack et al., 31 microduplication patients are characterised by behavioural problems and poor social interaction, J Med Genet, vol.46, pp.524-530, 2009.

G. Coppola, S. Chinnathambi, J. J. Lee, B. A. Dombroski, M. C. Baker et al., Evidence for a role of the rare p.A152T variant in MAPT in increasing the risk for FTD-spectrum and Alzheimer's diseases, Hum Mol Genet, vol.21, pp.3500-3512, 2012.

C. Duyckaerts, H. Braak, J. Brion, L. Buée, D. Tredici et al., PART is part of Alzheimer disease, Acta Neuropathol (Berl), vol.129, pp.7-11, 2015.

K. A. Jellinger, I. Alafuzoff, J. Attems, T. G. Beach, N. J. Cairns et al., PART, a distinct tauopathy, different from classical sporadic Alzheimer disease, Acta Neuropathol (Berl), vol.129, pp.757-762, 2015.

G. Jun, C. A. Ibrahim-verbaas, M. Vronskaya, J. Lambert, J. Chung et al., A novel Alzheimer disease locus located near the gene encoding tau protein, Mol Psychiatry, vol.21, pp.108-117, 2016.

D. References-backenroth, J. Homsy, L. R. Murillo, J. Glessner, E. Lin et al., CANOES: detecting rare copy number variants from whole exome sequencing data, Nucleic Acids Res, vol.42, p.97, 2014.

X. Bai, E. Rajendra, G. Yang, Y. Shi, and S. H. Scheres, Sampling the conformational space of the catalytic subunit of human ?-secretase, 2015.

L. Chávez-gutiérrez, A. Tolia, E. Maes, T. Li, P. C. Wong et al., Glu(332) in the Nicastrin ectodomain is essential for gamma-secretase complex maturation but not for its activity, J. Biol. Chem, vol.283, pp.20096-20105, 2008.

L. Chávez-gutiérrez, L. Bammens, I. Benilova, A. Vandersteen, M. Benurwar et al., The mechanism of ?-secretase dysfunction in familial Alzheimer disease, EMBO J, vol.31, pp.2261-2274, 2012.

A. Herreman, L. Serneels, W. Annaert, D. Collen, L. Schoonjans et al., Total inactivation of gamma-secretase activity in presenilin-deficient embryonic stem cells, Nat. Cell Biol, vol.2, pp.461-462, 2000.

M. Hiltunen, S. Helisalmi, A. Mannermaa, I. Alafuzoff, A. M. Koivisto et al., Identification of a novel 4.6-kb genomic deletion in presenilin-1 gene which results in exclu early onset Alzheimer's disease family: an Alu core sequence-stimulated recombination?, Eur. J. Hum. Genet, vol.8, pp.259-266, 2000.

N. Jurisch-yaksi, R. Sannerud, and W. Annaert, A fast growing spectrum of biological functions of ?-secretase in development and disease, Biochim. Biophys. Acta, vol.1828, pp.2815-2827, 2013.

H. Karlstrom, W. S. Brooks, J. B. Kwok, G. A. Broe, J. J. Kril et al., Variable phenotype of Alzheimer's disease with spastic paraparesis, J. Neurochem, vol.104, pp.573-583, 2008.

L. Guennec, K. Nicolas, G. Quenez, O. Charbonnier, C. Wallon et al., ABCA7 rare variants and Alzheimer disease risk, vol.86, pp.2134-2137, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01831744

L. Guennec, K. Quenez, O. Nicolas, G. Wallon, D. Rousseau et al., 17q21.31 duplication causes prominent tau-related dementia with increased MAPT expression, Mol. Psychiatry Lee, vol.3, pp.756-760, 1997.
URL : https://hal.archives-ouvertes.fr/hal-01832142

G. M. Mckhann, D. S. Knopman, H. Chertkow, B. T. Hyman, C. R. Jack et al., The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease, Alzheimers Dement. J. Alzheimers Assoc, vol.7, pp.263-269, 2011.

G. Nicolas, C. Charbonnier, D. Wallon, O. Quenez, C. Bellenguez et al., SORL1 rare variants: a major risk factor for familial early-onset Alzheimer's disease, Mol. Psychiatry, vol.21, pp.831-836, 2016.

G. Nicolas, D. Wallon, C. Charbonnier, O. Quenez, S. Rousseau et al., Screening of dementia genes by whole-exome sequencing in early-onset Alzheimer disease: input and lessons, Eur. J. Hum. Genet, vol.24, pp.710-716, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01431285

J. Perez-tur, S. Froelich, G. Prihar, R. Crook, M. Baker et al., A mutation in Alzheimer's disease destroying a splice acceptor site in the presenilin-1 gene, Neuroreport, vol.7, pp.297-301, 1995.

G. Prihar, A. Verkkoniem, J. Perez-tur, R. Crook, S. Lincoln et al., Alzheimer disease PS-1 exon 9 deletion defined, Nat. Med, vol.5, p.1090, 1999.

K. Rascovsky, J. R. Hodges, D. Knopman, M. F. Mendez, J. H. Kramer et al., Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia, Brain J. Neurol, vol.134, pp.2456-2477, 2011.

A. Rovelet-lecrux, D. Hannequin, G. Raux, N. Le-meur, A. Laquerrière et al., APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy, Nat. Genet, vol.38, pp.24-26, 2006.

A. Rovelet-lecrux, C. Charbonnier, D. Wallon, G. Nicolas, M. N. Seaman et al., De novo deleterious genetic variations target a biological network centered on A? peptide in early-onset Alzheimer disease, Mol. Psychiatry, vol.20, pp.1046-1056, 2015.

T. Saito, T. Suemoto, N. Brouwers, K. Sleegers, S. Funamoto et al., Potent amyloidogenicity and pathogenicity of A?43, Nat. Neurosci, vol.14, pp.1023-1032, 2011.

S. Sato, K. Kamino, T. Miki, A. Doi, K. Ii et al., Splicing mutation of presenilin-1 gene for early-onset familial Alzheimer's disease, Hum. Mutat. Suppl, vol.1, pp.91-94, 1998.

M. J. Smith, J. B. Kwok, C. A. Mclean, J. J. Kril, G. A. Broe et al., Variable phenotype of Alzheimer's disease with spastic paraparesis, Ann. Neurol, vol.49, pp.125-129, 2001.

H. Steiner, H. Romig, M. G. Grim, U. Philipp, B. Pesold et al., , p.1, 1999.

, Deltaexon 9 mutation is independent of its defect to undergo proteolytic processing, J. Biol. Chem, vol.274, pp.7615-7618

M. Szaruga, S. Veugelen, M. Benurwar, S. Lismont, D. Sepulveda-falla et al., Qualitative changes in human ?-secretase underlie familial Alzheimer's disease, J. Exp. Med, vol.212, pp.2003-2013, 2015.

M. Takami, Y. Nagashima, Y. Sano, S. Ishihara, M. Morishima-kawashima et al., gamma-Secretase: successive tripeptide and tetrapeptide release from the transmembrane domain of beta-carboxyl terminal fragment, J. Neurosci, vol.29, pp.13042-13052, 2009.

S. Veugelen, T. Saito, T. C. Saido, L. Chávez-gutiérrez, and B. De-strooper, Familial Alzheimer's disease mutations in presenilin generate amyloidogenic A? peptide seeds, Neuron, vol.90, pp.410-416, 2016.

J. Wanngren, J. Frånberg, A. I. Svensson, H. Laudon, F. Olsson et al., The large hydrophilic loop of presenilin 1 is important for regulating gamma-secretase complex assembly and dictating the amyloid beta peptide (Abeta) Profile without affecting Notch processing, Biol. Chem, vol.285, pp.8527-8536, 2010.

J. C. Lambert, C. A. Ibrahim-verbaas, D. Harold, A. C. Naj, R. Sims et al., Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease, Nat Genet, vol.45, issue.1, pp.5-7, 2013.

T. Jonsson, H. Stefansson, S. Steinberg, I. Jonsdottir, P. V. Jonsson et al., Variant of TREM2 associated with the risk of Alzheimer's disease, N Engl J Med, vol.368, pp.107-116, 2013.

C. Cruchaga, C. M. Karch, S. C. Jin, B. A. Benitez, Y. Cai et al., Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer's disease, Nature, vol.505, pp.550-554, 2014.

J. Gratten, P. M. Visscher, B. J. Mowry, and N. R. Wray, Interpreting the role of de novo protein-coding mutations in neuropsychiatric disease, Nat Genet, vol.45, pp.234-238, 2013.

J. A. Veltman and H. G. Brunner, De novo mutations in human genetic disease, Nat Rev Genet, vol.13, pp.565-575, 2012.

G. M. Mckhann, D. S. Knopman, H. Chertkow, B. T. Hyman, J. Cr et al., The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease, Alzheimers Dement, vol.7, issue.2, pp.3-269, 2011.

V. M. Perreau, S. Orchard, P. A. Adlard, S. A. Bellingham, R. Cappai et al., A domain level interaction network of amyloid precursor protein and Abeta of Alzheimer's disease, Proteomics, vol.10, pp.2377-95, 2010.

S. Gokool, D. Tattersall, and M. N. Seaman, EHD1 interacts with retromer to stabilize SNX1 tubules and facilitate endosome-to-Golgi retrieval, Traffic, vol.8, pp.1873-1886, 2007.

E. Helfer, M. E. Harbour, V. Henriot, G. Lakisic, C. Sousa-blin et al., Endosomal recruitment of the WASH complex: active sequences and mutations impairing interaction with the retromer, Biol Cell, vol.105, pp.191-207, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01960923

S. Y. Breusegem and M. N. Seaman, Image-based and biochemical assays to investigate endosomal protein sorting, Methods Enzymol, vol.534, pp.1-5, 2014.

M. E. Harbour, S. Y. Breusegem, R. Antrobus, C. Freeman, R. E. Seaman et al., The cargo-selective retromer complex is a recruiting hub for protein complexes that regulate endosomal tubule dynamics, J Cell Sci, vol.123, pp.3703-3717, 2010.

M. N. Seaman, M. E. Harbour, D. Tattersall, E. Read, and N. Bright, Membrane recruitment of the cargo-selective retromer subcomplex is catalysed by the small GTPase Rab7 and inhibited by the Rab-GAP TBC1D5, J Cell Sci, vol.122, pp.2371-2382, 2009.

D. Nesic, M. C. Miller, Z. T. Quinkert, M. Stein, B. T. Chait et al., Helicobacter pylori CagA inhibits PAR1-MARK family kinases by mimicking host substrates, Nat Struc Mol Biol, vol.17, pp.130-132, 2010.

T. A. Halgren, R. B. Murphy, R. A. Friesner, H. S. Beard, L. L. Frye et al., Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening, J Med Chem, vol.47, pp.1750-1759, 2004.

K. Lindorff-larsen, S. Piana, K. Palmo, P. Maragakis, J. L. Klepeis et al., Improved side-chain torsion potentials for the Amber ff99SB protein force field, Proteins, vol.78, pp.1950-1958, 2010.

C. D. Campbell and E. E. Eichler, Properties and rates of germline mutations in humans, Trends Genet, vol.29, pp.5-7, 2013.

J. Hardy and D. J. Selkoe, The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics, Science, vol.297, pp.3-5, 2002.

C. Pottier, D. Wallon, A. R. Lecrux, D. Maltete, S. Bombois et al., Amyloid-beta protein precursor gene expression in alzheimer's disease and other conditions, J Alzheimers Dis, vol.28, pp.561-566, 2012.

A. Rovelet-lecrux, D. Hannequin, G. Raux, L. Meur, N. Laquerriere et al., APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy, Nature Genet, vol.38, pp.2-4, 2006.

K. Sleegers, N. Brouwers, I. Gijselinck, J. Theuns, D. Goossens et al., APP duplication is sufficient to cause early onset Alzheimer's dementia with cerebral amyloid angiopathy, Brain, vol.129, pp.2977-2983, 2006.

A. K. Maunakea, R. P. Nagarajan, M. Bilenky, T. J. Ballinger, D. Souza et al., Conserved role of intragenic DNA methylation in regulating alternative promoters, Nature, vol.466, issue.2, pp.3-257, 2010.

. Encode-project-consortium, B. E. Bernstein, E. Birney, I. Dunham, E. D. Green et al., An integrated encyclopedia of DNA elements in the human genome, Nature, vol.489, pp.5-7, 2012.

S. O. Abdul-hay, T. Sahara, M. Mcbride, D. Kang, and M. A. Leissring, Identification of BACE2 as an avid ?-amyloid-degrading protease, Mol Neurodegener, vol.7, p.6, 2012.

X. Sun, G. He, and W. Song, BACE2 as a novel APP theta-secretase, is not responsible for the pathogenesis of Alzheimer's disease in Down syndrome, FASEB J, vol.20, pp.1369-1376, 2006.

J. Perez-tur, S. Froelich, G. Prihar, R. Crook, M. Baker et al., A mutation in Alzheimer's disease destroying a splice acceptor site in the presenilin-1 gene, Neuroreport, vol.7, issue.2, pp.7-301, 1995.

H. Steiner, H. Romig, M. G. Grim, U. Philipp, B. Pesold et al., The biological and pathological function of the presenilin-1 Deltaexon 9 mutation is independent of its defect to undergo proteolytic processing, J Biol Chem, vol.274, pp.7615-7618, 1999.

E. Rogaeva, Y. Meng, J. H. Lee, Y. Gu, T. Kawarai et al., The neuronal sortilinrelated receptor SORL1 is genetically associated with Alzheimer disease, Nat Genet, vol.39, pp.168-177, 2007.

C. Pottier, D. Hannequin, S. Coutant, A. Rovelet-lecrux, D. Wallon et al., High frequency of potentially pathogenic SORL1 mutations in autosomal dominant early-onset Alzheimer disease, Mol Psychiatry, vol.17, issue.8, pp.5-879, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00965204

M. N. Seaman, A. Gautreau, and D. D. Billadeau, Retromer-mediated endosomal protein sorting: all WASHed up!, Trends Cell Biol, vol.23, pp.5-7, 2013.

I. J. Mcgough, F. Steinberg, D. Jia, P. A. Barbuti, K. J. Mcmillan et al., Retromer binding to FAM21 and the WASH complex Is perturbed by the Parkinson diseaselinked VPS35(D620N) mutation, Curr Biol, vol.24, pp.1-6, 2014.

E. Zavodszky, M. N. Seaman, K. Moreau, M. Jimenez-sanchez, S. Y. Breusegem et al., Mutation in VPS35 associated with Parkinson's disease impairs WASH complex association and inhibits autophagy, Nat Commun, vol.5, p.3828, 2014.

S. A. Small, K. Kent, A. Pierce, C. Leung, M. S. Kang et al., Model-guided microarray implicates the retromer complex in Alzheimer's disease, Ann Neurol, vol.58, pp.909-919, 2005.

A. Muhammad, I. Flores, H. Zhang, R. Yu, A. Staniszewski et al., Retromer deficiency observed in Alzheimer's disease causes hippocampal dysfunction, neurodegeneration, and Abeta accumulation, Proc Natl Acad Sci, vol.105, pp.7327-7332, 2008.

C. P. Sullivan, A. G. Jay, E. C. Stack, M. Pakaluk, E. Wadlinger et al., Retromer disruption promotes amyloidogenic APP processing, Neurobiol Dis, vol.43, pp.338-345, 2011.

L. Wen, F. L. Tang, Y. Hong, S. W. Luo, C. L. Wang et al., VPS35 haploinsufficiency increases Alzheimer's disease neuropathology, J Cell Biol, vol.195, pp.765-779, 2011.

A. Bhalla, C. P. Vetanovetz, E. Morel, Z. Chamoun, D. Paolo et al., The location and trafficking routes of the neuronal retromer and its role in amyloid precursor protein transport, Neurobiol Dis, vol.47, pp.1-2, 2012.

R. W. Choy, Z. Cheng, and R. Schekman, Amyloid precursor protein (APP) traffics from the cell surface via endosomes for amyloid beta (Abeta) production in the trans-Golgi network, Proc Natl Acad Sci, vol.109, pp.2077-2082, 2012.

R. F. Lane, S. M. Raines, J. W. Steele, M. E. Ehrlich, J. A. Lah et al., Diabetesassociated SorCS1 regulates Alzheimer's amyloid-beta metabolism: evidence for involvement of SorL1 and the retromer complex, J Neurosci, vol.30, pp.13110-13115, 2010.

S. I. Vieira, S. Rebelo, H. Esselmann, J. Wiltfang, J. Lah et al., Retrieval of the Alzheimer's amyloid precursor protein from the endosome to the TGN is S655 phosphorylation state-dependent and retromer-mediated, Mol Neurodegener, vol.5, p.40, 2010.

A. W. Fjorback, M. Seaman, C. Gustafsen, A. Mehmedbasic, S. Gokool et al., Retromer binds the FANSHY sorting motif in SorLA to regulate amyloid precursor protein sorting and processing, J Neurosci, vol.32, pp.1467-1480, 2012.

V. J. Mecozzi, D. E. Berman, S. Simoes, C. Vetanovetz, M. R. Awal et al., Pharmacological chaperones stabilize retromer to limit APP processing, Nat Chem Biol, vol.10, pp.443-449, 2014.

G. Drewes, MARKing tau for tangles and toxicity, Trends Biochem Sci, vol.29, pp.548-555, 2004.

G. J. Gu, H. Lund, D. Wu, A. Blokzijl, C. Classon et al., Role of individual MARK isoforms in phosphorylation of tau at Ser(2)(6)(2) in Alzheimer's disease, Neuromol Med, vol.15, pp.4-5, 2013.

W. Yu, J. Polepalli, D. Wagh, J. Rajadas, R. Malenka et al., A critical role for the PAR-1/ MARK-tau axis in mediating the toxic effects of Abeta on synapses and dendritic spines, Hum Mol Genet, vol.21, pp.1-3, 2012.

K. Iijima, A. Gatt, and K. Iijima-ando, Tau Ser262 phosphorylation is critical for Abeta42induced tau toxicity in a transgenic Drosophila model of Alzheimer's disease, Hum Mol Genet, vol.19, pp.2947-2957, 2010.

I. Nishimura, Y. Yang, and B. Lu, PAR-1 kinase plays an initiator role in a temporally ordered phosphorylation process that confers tau toxicity in Drosophila, Cell, vol.116, pp.6-7, 2004.

F. Naz, F. Anjum, A. Islam, F. Ahmad, and M. I. Hassan, Microtubule affinity-regulating kinase 4: structure, function, and regulation, Cell Biochem Biophys, vol.67, pp.485-499, 2013.

T. Tanoue, M. Adachi, T. Moriguchi, and E. Nishida, A conserved docking motif in MAP kinases common to substrates, activators and regulators, Nat Cell Biol, vol.2, pp.110-116, 2000.

I. Iossifov, B. J. O'roak, S. J. Sanders, M. Ronemus, N. Krumm et al., The contribution of de novo coding mutations to autism spectrum disorder, Nature, vol.515, issue.2, pp.1-6, 2014.

S. De-rubeis, X. He, A. P. Goldberg, C. S. Poultney, K. Samocha et al., Synaptic, transcriptional and chromatin genes disrupted in autism, Nature, vol.515, pp.209-215, 2014.

I. Iossifov, M. Ronemus, D. Levy, Z. Wang, I. Hakker et al., De novo gene disruptions in children on the autistic spectrum, Neuron, vol.74, issue.2, pp.5-299, 2012.

B. M. Neale, Y. Kou, L. Liu, A. Ma'ayan, K. E. Samocha et al., Patterns and rates of exonic de novo mutations in autism spectrum disorders, Nature, vol.485, pp.242-245, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00939274

C. Dumanchin, A. Brice, D. Campion, D. Hannequin, C. Martin et al., De novo presenilin 1 mutations are rare in clinically sporadic, early onset Alzheimer's disease cases. French Alzheimer's Disease Study Group, J Med Genet, vol.35, pp.672-673, 1998.

F. Portet, Y. Dauvilliers, D. Campion, G. Raux, J. J. Hauw et al., Very early onset AD with a de novo mutation in the presenilin 1 gene (Met 233 Leu), Neurology, vol.61, pp.1136-1137, 2003.

M. P. Golan, M. Styczy?ska, K. Jó?wiak, J. Walecki, A. Maruszak et al., Earlyonset Alzheimer's disease with a de novo mutation in the presenilin 1 gene, Exp Neurol, vol.208, pp.264-268, 2007.

L. E. Vissers, J. De-ligt, C. Gilissen, I. Janssen, M. Steehouwer et al., A de novo paradigm for mental retardation, Nat Genet, vol.42, pp.1109-1112, 2010.

G. R. Abecasis, A. Auton, L. D. Brooks, M. A. Depristo, R. M. Durbin et al., An integrated map of genetic variation from 1,092 human genomes, Genomes Project Consortium, vol.491, pp.56-65, 2012.

. Genomes-project-consortium, A. Auton, L. D. Brooks, R. M. Durbin, E. P. Garrison et al., A global reference for human genetic variation, Nature, vol.526, pp.68-74, 2015.

R. Acuna-hidalgo, J. A. Veltman, and A. Hoischen, New insights into the generation and role of de novo mutations in health and disease, Genome Biol, vol.17, p.241, 2016.

I. Adzhubei, D. M. Jordan, and S. R. Sunyaev, Predicting functional effect of human missense mutations using PolyPhen-2, Curr. Protoc. Hum. Genet. Chapter, p.20, 2013.

J. Alexander, O. Kalev, S. Mehrabian, L. Traykov, M. Raycheva et al., Familial early-onset dementia with complex neuropathologic phenotype and genomic background, Neurobiol. Aging, vol.42, pp.199-204, 2016.

C. Alkan, B. P. Coe, and E. E. Eichler, Genome structural variation discovery and genotyping, Nat. Rev. Genet, vol.12, pp.363-376, 2011.

A. Alzheimer, R. A. Stelzmann, H. N. Schnitzlein, and F. R. Murtagh, An English translation of Alzheies pape, Ue eie eigeaatige Ekaakug de Hiide, Cli. Aat. N. Y. N, vol.8, pp.429-431, 1995.

O. M. Andersen, J. Reiche, V. Schmidt, M. Gotthardt, R. Spoelgen et al., Neuronal sorting protein-related receptor sorLA/LR11 regulates processing of the amyloid precursor protein, Proc. Natl. Acad. Sci. U. S. A, vol.102, pp.13461-13466, 2005.

O. M. Andersen, I. Rudolph, and T. E. Willnow, Risk factor SORL1: from genetic association to futioal alidatio i Alzheies disease, Ata Neuopathol. Bel, vol.132, pp.653-665, 2016.

D. Backenroth, J. Homsy, L. R. Murillo, J. Glessner, E. Lin et al., CANOES: detecting rare copy number variants from whole exome sequencing data, Nucleic Acids Res, vol.42, p.97, 2014.

X. Bai, C. Yan, G. Yang, P. Lu, D. Ma et al., An atomic structure of human ?-secretase, Nature, vol.525, pp.212-217, 2015.

I. J. Barnett, S. Lee, L. , and X. , Detecting rare variant effects using extreme phenotype sampling in sequencing association studies, Genet. Epidemiol, vol.37, pp.142-151, 2013.

E. C. Beattie, R. C. Carroll, X. Yu, W. Morishita, H. Yasuda et al., Regulation of AMPA receptor endocytosis by a signaling mechanism shared with LTD, Nat. Neurosci, vol.3, pp.1291-1300, 2000.

I. Benilova, E. Karran, D. Strooper, and B. , The toxic A? oligoe aad Alzheies disease: aa emperor in need of clothes, Nat. Neurosci, vol.15, pp.349-357, 2012.

S. T. Bennett, C. Barnes, A. Cox, L. Davies, and C. Brown, Toward the 1,000 dollars human genome, Pharmacogenomics, vol.6, pp.373-382, 2005.

G. S. Bloom, Amyloid-? and tau: the trigger and bullet in Alzheimer disease pathogenesis, JAMA Neurol, vol.71, pp.505-508, 2014.

B. Boucher, J. , and S. , Genetic interaction networks: better understand to better predict, p.290, 2013.

H. Braak and E. Braak, Neuropathological stageing of Alzheimer-related changes, Acta Neuropathol. (Berl.), vol.82, pp.239-259, 1991.

N. Brouwers, C. Van-cauwenberghe, S. Engelborghs, J. Lambert, K. Bettens et al., Alzheimer risk associated with a copy number variation in the complement receptor 1 increasing C3b/C4b binding sites, Mol. Psychiatry, vol.17, pp.223-233, 2012.

R. Cacace, T. Van-den-bossche, S. Engelborghs, N. Geerts, A. Laureys et al., Rare Variants in PLD3 Do Not Affect Risk for Early-Onset Alzheimer Disease in a European Consortium Cohort, Hum. Mutat, vol.36, pp.1226-1235, 2015.

R. Cacace, K. Sleegers, and C. Van-broeckhoven, Molecular genetics of early-onset Alzheies disease eisited, Alzheies Deet. J. Alzheies Asso, vol.12, pp.733-748, 2016.

S. Caglayan, S. Takagi-niidome, F. Liao, A. Carlo, V. Schmidt et al., Lysosomal sorting of amyloid-? by the SORLA eepto is ipaied a failial Alzheies disease utatio, 2014.

X. D. Cai, T. E. Golde, and S. G. Younkin, Release of excess amyloid beta protein from a mutant amyloid beta protein precursor, Science, vol.259, pp.514-516, 1993.

A. De-calignon, T. L. Spires-jones, R. Pitstick, G. A. Carlson, H. et al., Tangle-bearing neurons survive despite disruption of membrane integrity in a mouse model of tauopathy, J. Neuropathol. Exp. Neurol, vol.68, pp.757-761, 2009.

C. D. Campbell and E. E. Eichler, Properties and rates of germline mutations in humans, Trends Genet. TIG, vol.29, pp.575-584, 2013.

M. Cargill, D. Altshuler, J. Ireland, P. Sklar, K. Ardlie et al., Characterization of single-nucleotide polymorphisms in coding regions of human genes, Nat. Genet, vol.22, pp.231-238, 1999.

C. M. Carvalho and J. R. Lupski, Mechanisms underlying structural variant formation in genomic disorders, Nat. Rev. Genet, vol.17, pp.224-238, 2016.

S. Castellana and T. Mazza, Congruency in the prediction of pathogenic missense mutations: state-of-the-art web-based tools, Brief. Bioinform, vol.14, pp.448-459, 2013.

M. J. Chaisson, R. K. Wilson, and E. E. Eichler, Genetic variation and the de novo assembly of human genomes, Nat. Rev. Genet, vol.16, pp.627-640, 2015.

J. Chapman, E. Rees, D. Harold, D. Ivanov, A. Gerrish et al., A genome-wide study shows a limited contribution of aae op ue aaiaats to Alzheies disease isk, Hum. Mol. Genet, vol.22, pp.816-824, 2013.

J. Chapuis, A. Flaig, B. Grenier-boley, F. Eysert, V. Pottiez et al., Genome-wide, high-content siRNA screening identifies the Alzheies geeti isk faato FE'MT as a ajo odulato of APP etaaolis, Ata Neuopathol, 2016.

L. Chávez-gutiérrez, L. Bammens, I. Benilova, A. Vandersteen, M. Benurwar et al., The mechanism of ?-Secretase dysfunction in familial Alzheimer disease, EMBO J, vol.31, pp.2261-2274, 2012.

C. Chiang, A. J. Scott, J. R. Davis, E. K. Tsang, X. Li et al., The impact of structural variation on human gene expression, Nat. Genet, 2017.

M. Citron, T. Oltersdorf, C. Haass, L. Mcconlogue, A. Y. Hung et al., Mutation of the beta-amyloid precursor protein in familial Alzheies disease ieases eta-protein production, Nature, vol.360, pp.672-674, 1992.

M. Citron, D. Westaway, W. Xia, G. Carlson, T. Diehl et al., Mutaat peseilis of Alzheies disease iease podutio of 42-residue amyloid beta-protein in both transfected cells and transgenic mice, Nat. Med, vol.3, pp.67-72

D. W. Cleveland, S. Y. Hwo, and M. W. Kirschner, Purification of tau, a microtubule-associated protein that induces assembly of microtubules from purified tubulin, J. Mol. Biol, vol.116, pp.207-225, 1977.

B. P. Coe, K. Witherspoon, J. A. Rosenfeld, B. W. Van-bon, A. T. Vulto-van-silfhout et al., Refining analyses of copy number variation identifies specific genes associated with developmental delay, Nat. Genet, vol.46, pp.1063-1071, 2014.

C. Condello, P. Yuan, A. Schain, and J. Grutzendler, Microglia constitute a barrier that prevents neurotoxic protofibrillar A?42 hotspots around plaques, Nat. Commun, vol.6, p.6176, 2015.

D. F. Conrad, D. Pinto, R. Redon, L. Feuk, O. Gokcumen et al., Origins and functional impact of copy number variation in the human genome, Nature, vol.464, pp.704-712, 2010.

J. F. Crary, J. Q. Trojanowski, J. A. Schneider, J. F. Abisambra, E. L. Abner et al., Primary age-related tauopathy (PART): a common pathology associated with human aging, Acta Neuropathol. (Berl.), vol.128, pp.755-766, 2014.

R. Crook, A. Verkkoniemi, J. Perez-tur, N. Mehta, M. Baker et al., A variant of Alzheies disease ith spasti paapaaesis aad unusual plaques due to deletion of exon 9 of presenilin 1, Nat. Med, vol.4, pp.452-455, 1998.

C. Cruchaga, C. M. Karch, S. C. Jin, B. A. Benitez, Y. Cai et al., Rare coding variants in the phospholipase D3 gene confer risk for Alzheies disease, Natue, vol.505, pp.550-554, 2014.

E. Cues and K. Leeges, Geeti aaiatios udelig Alzheies disease: eidee fo genome-wide association studies and beyond, Lancet Neurol, vol.15, pp.857-868

E. Cuyvers, A. De-roeck, T. Van-den-bossche, C. Van-cauwenberghe, K. Bettens et al., Mutations in ABCA7 i a Belgiaa ohot of Alzheies disease patiets: a taageted eseueig stud, vol.14, pp.814-822, 2015.

H. N. Dawson, A. Ferreira, M. V. Eyster, N. Ghoshal, L. I. Binder et al., Inhibition of neuronal maturation in primary hippocampal neurons from tau deficient mice, J. Cell Sci, vol.114, pp.1179-1187, 2001.

C. De-jonghe, C. Esselens, S. Kumar-singh, K. Craessaerts, S. Serneels et al., Pathogenic APP mutations near the gamma-secretase cleavage site differentially affect Abeta secretion and APP C-terminal fragment stability, Hum. Mol. Genet, vol.10, pp.1665-1671, 2001.

B. De-strooper, P. Saftig, K. Craessaerts, H. Vanderstichele, G. Guhde et al., Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein, Nature, vol.391, pp.387-390, 1998.

A. Delacourte, J. P. David, N. Sergeant, L. Buée, A. Wattez et al., The biochemical pathway of neurofibrillary degeneration in agig aad Alzheies disease, Neuolog, vol.52, pp.1158-1165, 1999.

C. Delay, B. Grenier-boley, P. Amouyel, J. Dumont, L. et al., miRNA-dependent target regulation: functional characterization of single-nucleotide polymorphisms identified in genome-ide assoiatio studies of Alzheies disease, Alzheies 'es. The, vol.8, p.20, 2016.

F. E. Dee, M. F. Mua, J. D. Oeto, L. Haaegge, J. B. Leade et al., Distribution and clinical impact of functional variants in 50,726 whole-exome sequences from the DiscovEHR study, Science, vol.354, 2016.

S. E. Dodson, O. M. Andersen, V. Karmali, J. J. Fritz, D. Cheng et al., Loss of LR11/SORLA enhances early pathology in a mouse model of amyloidosis: evidence fo a poial ole i Alzheies disease, J. Neuosi. Off. J. "o. Neuosi, vol.28, pp.12877-12886, 2008.

C. Dong, P. Wei, X. Jian, R. Gibbs, E. Boerwinkle et al., Comparison and integration of deleteriousness prediction methods for nonsynonymous SNVs in whole exome sequencing studies, Hum. Mol. Genet, vol.24, pp.2125-2137, 2015.

P. Dourlen, F. J. Fernandez-gomez, C. Dupont, B. Grenier-boley, C. Bellenguez et al., Functional screening of Alzheimer risk loci identifies PTK2B as an in vivo modulator and early marker of Tau pathology, Mol. Psychiatry, 2016.

C. Dumanchin, I. Tournier, C. Martin, M. Didic, S. Belliard et al., Biological effects of four PSEN1 gene mutations causing Alzheimer disease with spastic paraparesis and cotton wool plaques, Hum. Mutat, vol.27, p.1063, 2006.

S. B. Dumanis, T. Burgert, S. Caglayan, A. Füchtbauer, E. Füchtbauer et al., Distinct Functions for Anterograde and Retrograde Sorting of SORLA in Amyloidogenic Processes in the Brain, J. Neurosci. Off. J. Soc. Neurosci, vol.35, pp.12703-12713, 2015.

C. Duyckaerts, J. J. Hauw, F. Piette, C. Rainsard, V. Poulain et al., , 1985.

, Cortical atrophy in senile dementia of the Alzheimer type is mainly due to a decrease in cortical length, Acta Neuropathol. (Berl.), vol.66, pp.72-74

C. Duyckaerts, M. A. Colle, B. Delatour, and J. J. Hauw,

, Rev. Neurol. (Paris), vol.155, pp.17-27

M. D. Ehlers, Reinsertion or degradation of AMPA receptors determined by activitydependent endocytic sorting, Neuron, vol.28, pp.511-525, 2000.

H. Englund, D. Sehlin, A. Johansson, L. N. Nilsson, P. Gellerfors et al., Sensitive ELISA detection of amyloid-beta protofibrils in biological samples, J. Neurochem, vol.103, pp.334-345, 2007.

M. Espinoza, R. De-silva, D. W. Dickson, and P. Davies, Differential incorporation of tau isofos i Alzheies disease, J. Alzheies Dis. JAD, vol.14, pp.1-16, 2008.

P. Fazzari, K. Horre, A. M. Arranz, C. S. Frigerio, T. Saito et al., PLD3 gene and processing of APP, Nature, vol.541, pp.1-2, 2017.

H. V. Firth, S. M. Richards, A. P. Bevan, S. Clayton, M. Corpas et al., DECIPHER: Database of Chromosomal Imbalance and Phenotype in Humans Using Ensembl Resources, Am. J. Hum. Genet, vol.84, pp.524-533, 2009.

J. Folch, D. Petrov, M. Ettcheto, S. Abad, E. Sánchez-lópez et al., Current Research Therapeutic Strategies for Alzheies Disease Teatet, Neual Plast, p.8501693, 2016.

M. L. Frandemiche, S. De-seranno, T. Rush, E. Borel, A. Elie et al., Activity-dependent tau protein translocation to excitatory synapse is disrupted by exposure to amyloid-beta oligomers, J. Neurosci. Off. J. Soc. Neurosci, vol.34, pp.6084-6097, 2014.

M. Fromer, J. L. Moran, K. Chambert, E. Banks, S. E. Bergen et al., Discovery and statistical genotyping of copy-number variation from whole-exome sequencing depth, Am. J. Hum. Genet, vol.91, pp.597-607, 2012.

M. Fromer, A. J. Pocklington, D. H. Kavanagh, H. J. Williams, S. Dwyer et al., De novo mutations in schizophrenia implicate synaptic networks, Nature, vol.506, pp.179-184, 2014.

Y. Fu, J. T. Hsiao, G. Paxinos, G. M. Halliday, K. et al., ABCA7 Mediates Phagocytic Clearance of Amyloid-? in the Brain, J. Alzheimers Dis. JAD, vol.54, pp.569-584, 2016.

K. Fujio, M. Sato, T. Uemura, T. Sato, R. Sato-harada et al., 14-3-3 proteins and protein phosphatases are not reduced in tau-deficient mice, Neuroreport, vol.18, pp.1049-1052, 2007.

P. Garagnani, C. Giuliani, C. Pirazzini, F. Olivieri, M. G. Bacalini et al., Centenarians as super-controls to assess the biological relevance of genetic risk factors for common age-related diseases: a proof of principle on type 2 diabetes, Aging, vol.5, pp.373-385, 2013.

E. Genin, D. Hannequin, D. Wallon, K. Sleegers, M. Hiltunen et al., APOE and Alzheimer disease: a major gene with semi-dominant inheritance, Mol. Psychiatry, vol.16, pp.903-907, 2011.
DOI : 10.1038/mp.2011.52

URL : https://www.nature.com/articles/mp201152.pdf

G. Genovese, M. Fromer, E. A. Stahl, D. M. Ruderfer, K. Chambert et al., Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia, Nat. Neurosci, vol.19, pp.1433-1441, 2016.

G. Gibson, Rare and common variants: twenty arguments, Nat. Rev. Genet, vol.13, pp.135-145, 2012.
DOI : 10.1038/nrg3118

URL : http://europepmc.org/articles/pmc4408201?pdf=render

G. G. Glee and C. W. Wog, Alzheies disease: iitial epot of the puifiation and characterization of a novel cerebrovascular amyloid protein, Biochem. Biophys. Res. Commun, vol.120, pp.885-890

K. Goh, M. E. Cusick, D. Valle, B. Childs, M. Vidal et al., The human disease network, Proc. Natl. Acad. Sci. U. S. A, vol.104, pp.8685-8690, 2007.

A. González-pérez and N. López-bigas, Improving the assessment of the outcome of nonsynonymous SNVs with a consensus deleteriousness score, Condel, Am. J. Hum. Genet, vol.88, pp.440-449, 2011.

J. P. Greenfield, J. Tsai, G. K. Gouras, B. Hai, G. Thinakaran et al., Endoplasmic reticulum and trans-Golgi network generate distinct populations of Alzheimer beta-amyloid peptides, Proc. Natl. Acad. Sci. U. S. A, vol.96, pp.742-747, 1999.

R. Guerreiro, A. Wojtas, J. Bras, M. Carrasquillo, E. Rogaeva et al.,

, EM aaiaats i Alzheies disease. N. Egl. J. Med, vol.368, pp.117-127

R. J. Guerreiro, M. Baquero, R. Blesa, M. Boada, J. M. Brás et al., Geeti seeig of Alzheies disease gees i Ieiaa aad African samples yields novel mutations in presenilins and APP, Neurobiol. Aging, vol.31, pp.725-731

G. Guffanti, F. Torri, J. Rasmussen, A. P. Clark, A. Lakatos et al., ADNI the Alzheies Disease Neuoiagig Iitiatie

, Ieased CNVregion deletions in mild cognitive impairment (MCI) ad Alzheies disease AD sujets i the ADNI sample, Genomics, vol.102, pp.112-122

S. Guntupalli, J. Widagdo, A. , and V. , Amyloid-?-Induced Dysregulation of AMPA Receptor Trafficking, Neural Plast, p.3204519, 2016.

Y. Guo, Q. Sheng, D. C. Samuels, B. Lehmann, J. A. Bauer et al., Comparative study of exome copy number variation estimation tools using array comparative genomic hybridization as control, BioMed Res. Int, p.915636, 2013.

C. Haass, C. Kaether, G. Thinakaran, and S. Sisodia, Trafficking and proteolytic processing of APP. Cold Spring Harb, Perspect. Med, vol.2, p.6270, 2012.

A. Harada, K. Oguchi, S. Okabe, J. Kuno, S. Terada et al., Altered microtubule organization in small-calibre axons of mice lacking tau protein, Nature, vol.369, pp.488-491, 1994.

J. A. Haad and G. A. Higgis, Alzheies disease: the aloid asade hpothesis, vol.256, pp.184-185

D. Harold, R. Abraham, P. Hollingworth, R. Sims, A. Gerrish et al., , 2009.

, Genome-wide association study identifies aaiaats at CLU aad PICALM assoiated ith Alzheies disease, Nat. Geet, vol.41, pp.1088-1093

T. Hart, M. Chandrashekhar, M. Aregger, Z. Steinhart, K. R. Brown et al., High-Resolution CRISPR Screens Reveal Fitness Genes and Genotype-Specific Cancer Liabilities, Cell, vol.163, pp.1515-1526, 2015.

J. A. Hartigan, J. , and G. V. , Transient increases in intracellular calcium result in prolonged site-selective increases in Tau phosphorylation through a glycogen synthase kinase 3betadependent pathway, J. Biol. Chem, vol.274, pp.21395-21401, 1999.

T. Hartmann, S. C. Bieger, B. Brühl, P. J. Tienari, N. Ida et al., Distit sites of ittaaellulaa podutio fo Alzheies disease A beta40/42 amyloid peptides, Nat. Med, vol.3, pp.1016-1020

P. J. Hastings, J. R. Lupski, S. M. Rosenberg, I. , and G. , Mechanisms of change in gene copy number, Nat. Rev. Genet, vol.10, pp.551-564, 2009.

K. He, L. Song, L. W. Cummings, J. Goldman, R. L. Huganir et al., Stabilization of Ca2+-permeable AMPA receptors at perisynaptic sites by GluR1-S845 phosphorylation, Proc. Natl. Acad. Sci. U. S. A, vol.106, 2009.
DOI : 10.1073/pnas.0910338106

URL : http://www.pnas.org/content/106/47/20033.full.pdf

J. Y. Hehir-kwa, R. Pfundt, and J. A. Veltman, Exome sequencing and whole genome sequencing for the detection of copy number variation, Expert Rev. Mol. Diagn, vol.15, pp.1023-1032, 2015.

S. Heilmann, D. Drichel, J. Clarimon, V. Fernández, A. Lacour et al., PLD3 in non-failial Alzheies disease, vol.520, pp.3-5, 2015.
DOI : 10.1038/nature14039

K. Henjum, I. S. Almdahl, V. Årskog, L. Minthon, O. Hansson et al., Ceeospial fluid solule T'EM i agig aad Alzheies disease. Alzheies 'es. The. 8, p.17, 2016.

D. G. Hert, C. P. Fredlake, and A. E. Barron, Advantages and limitations of next-generation sequencing technologies: a comparison of electrophoresis and non-electrophoresis methods, Electrophoresis, vol.29, pp.4618-4626, 2008.

A. Heslegrave, W. Heywood, R. Paterson, N. Magdalinou, J. Svensson et al., Increased cerebrospinal fluid soluble TREM2 oetatio i Alzheies disease, Mol. Neuodegee, vol.11, p.3, 2016.
DOI : 10.1186/s13024-016-0071-x

URL : https://molecularneurodegeneration.biomedcentral.com/track/pdf/10.1186/s13024-016-0071-x?site=molecularneurodegeneration.biomedcentral.com

D. Hirtz, D. J. Thurman, K. Gwinn-hardy, M. Mohamed, A. R. Chaudhuri et al., , 2007.

, Ho oo aae the oo euologi disodes? Neuolog 68, pp.326-337

P. Hollingworth, D. Harold, R. Sims, A. Gerrish, J. Lambert et al., Common variants at ABCA7, M"AA/M"AE, EPHA, CD aad CDAP aae assoiated ith Alzheies disease, vol.43, pp.429-435, 2011.

B. B. Holmes and M. I. Diamond, Prion-like properties of Tau protein: the importance of extracellular Tau as a therapeutic target, J. Biol. Chem, vol.289, pp.19855-19861, 2014.

B. V. Hooli, Z. M. Kovacs-vajna, K. Mullin, M. A. Blumenthal, M. Mattheisen et al., Rare autosomal copy number variations in earlyonset familial Alzheies disease, Mol. Pshiatt, vol.19, pp.676-681, 2014.

B. V. Hooli, C. M. Lill, K. Mullin, D. Qiao, C. Lange et al., PLD3 gene aaiaats aad Alzheies disease, vol.520, pp.7-8, 2015.

C. Houdayer, V. Caux-moncoutier, S. Krieger, M. Barrois, F. Bonnet et al., Guidelines for splicing analysis in molecular diagnosis derived from a set of 327 combined in silico/in vitro studies on BRCA1 and BRCA2 variants, Hum. Mutat, vol.33, pp.1228-1238, 2012.

N. Huang, I. Lee, E. M. Marcotte, and M. E. Hurles, Characterising and predicting haploinsufficiency in the human genome, PLoS Genet, vol.6, 2010.

Y. A. Huang, B. Zhou, M. Wernig, and T. C. Südhof, ApoE2, ApoE3, and ApoE4 Differentially Stimulate APP Transcription and A? Secretion, vol.168, pp.427-441, 2017.

A. J. Iafrate, L. Feuk, M. N. Rivera, M. L. Listewnik, P. K. Donahoe et al., Detection of large-scale variation in the human genome, Nat. Genet, vol.36, pp.949-951, 2004.

A. Consortium, D. M. Gibbs, R. A. Peltonen, L. Altshuler, D. M. Gibbs et al., Integrating common and rare genetic variation in diverse human populations, Nature, vol.467, pp.52-58, 2010.

, Finishing the euchromatic sequence of the human genome, International Human Genome Sequencing Consortium, vol.431, pp.931-945, 2004.

I. Iossifo, B. J. O'oak, S. J. Sanders, M. Ronemus, N. Krumm et al., The contribution of de novo coding mutations to autism spectrum disorder, Nature, vol.515, pp.216-221, 2014.

C. R. Jack, D. S. Knopman, G. Chételat, D. Dickson, A. M. Fagan et al., Suspected non-Alzheimer disease pathophysiologyconcept and controversy, Nat. Rev. Neurol, vol.12, pp.117-124, 2016.

T. R. Jay, C. M. Miller, P. J. Cheng, L. C. Graham, S. Bemiller et al., TREM2 deficiency eliminates TREM2+ inflammatory aaophages aad aelioates patholog i Alzheies disease mouse models, J. Exp. Med, vol.212, pp.287-295, 2015.

X. Jian, E. Boerwinkle, and X. Liu, In silico prediction of splice-altering single nucleotide variants in the human genome, Nucleic Acids Res, vol.42, pp.13534-13544, 2014.

S. C. Jin, B. A. Benitez, C. M. Karch, B. Cooper, T. Skorupa et al., Codig aaiaats i T'EM iease isk fo Alzheies disease, Hu. Mol. Genet, vol.23, pp.5838-5846

T. Jonsson, J. K. Atwal, S. Steinberg, J. Snaedal, P. V. Jonsson et al., A utatio i APP potets agaist Alzheies disease and age-related cognitive decline, Nature, vol.488, pp.96-99

T. Jonsson, H. Stefansson, S. Steinberg, I. Jonsdottir, P. V. Jonsson et al., Variant of TREM2 associated with the risk of Alzheies disease, N. Egl. J. Med, vol.368, pp.107-116, 2013.

G. Jun, C. A. Ibrahim-verbaas, M. Vronskaya, J. Lambert, J. Chung et al., A novel Alzheimer disease locus located near the gene encoding tau protein, Mol. Psychiatry, vol.21, pp.108-117, 2016.

L. Kadalayil, S. Rafiq, M. J. Rose-zerilli, R. J. Pengelly, H. Parker et al., Exome sequence read depth methods for identifying copy number changes, Brief. Bioinform, vol.16, pp.380-392, 2015.

T. Kawarabayashi, M. Shoji, L. H. Younkin, L. Wen-lang, D. W. Dickson et al., Dimeric amyloid beta protein rapidly accumulates in lipid rafts followed by apolipoprotein E and phosphorylated tau accumulation in the Tg2576 mouse model of Alzheies disease, J. Neuosi. Off. J. "o. Neuosi, vol.24, pp.3801-3809, 2004.

R. Kayed, E. Head, J. L. Thompson, T. M. Mcintire, S. C. Milton et al., Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis, Science, vol.300, pp.486-489, 2003.

R. J. Kelleher and J. Shen, Genetics. Gamma-secretase and human disease, Science, vol.330, pp.1055-1056, 2010.

W. S. Kim, G. J. Guillemin, E. N. Glaros, C. K. Lim, and B. Garner, Quantitation of ATP-binding cassette subfamily-A transporter gene expression in primary human brain cells, Neuroreport, vol.17, pp.891-896, 2006.

W. S. Kim, H. Li, K. Ruberu, S. Chan, D. A. Elliott et al., Deletion of Abca7 increases cerebral amyloid-? accuulatio i the J ouse odel of Alzheies disease, J. Neurosci. Off. J. Soc. Neurosci, vol.33, pp.4387-4394, 2013.

M. Kihe, D. M. Witte, P. Jai, B. J. O'oak, G. M. Coope et al., A geeal framework for estimating the relative pathogenicity of human genetic variants, Nat. Genet, vol.46, pp.310-315

G. Kirov, A. J. Pocklington, P. Holmans, D. Ivanov, M. Ikeda et al., De novo CNV analysis implicates specific abnormalities of postsynaptic signalling complexes in the pathogenesis of schizophrenia, Mol. Psychiatry, vol.17, pp.142-153, 2012.

G. Kleinberger, Y. Yamanishi, M. Suárez-calvet, E. Czirr, E. Lohmann et al., TREM2 mutations implicated in neurodegeneration impair cell surface transport and phagocytosis, Sci. Transl. Med, vol.6, pp.243-86, 2014.

R. Koldamova, N. F. Fitz, and I. Lefterov, The role of ATP-binding cassette transporter A1 in Alzheies disease aad euodegeeatio, Biochim. Biophys. Acta, vol.1801, pp.824-830, 2010.

A. Kong, M. L. Frigge, G. Masson, S. Besenbacher, P. Sulem et al., Rate of de novo mutations and the ipotaae of fathes age to disease isk, Natue, vol.488, pp.471-475, 2012.

D. A. Koolen, L. E. Vissers, R. Pfundt, N. De-leeuw, S. J. Knight et al., A new chromosome 17q21.31 microdeletion syndrome associated with a common inversion polymorphism, Nat. Genet, vol.38, pp.999-1001, 2006.

D. A. Koolen, J. M. Kramer, K. Neveling, W. M. Nillesen, H. L. Moore-barton et al., Mutations in the chromatin modifier gene KANSL1 cause the 17q21.31 microdeletion syndrome, Nat. Genet, vol.44, pp.639-641, 2012.

G. G. Kovacs, Invited review: Neuropathology of tauopathies: principles and practice, Neuropathol. Appl. Neurobiol, vol.41, pp.3-23, 2015.

N. Ku, P. H. Ko, A. O'oak, B. J. Malig, M. Coe et al., Copy number variation detection and genotyping from exome sequence data, Genome Res, vol.22, pp.1525-1532, 2012.

P. Kuhn, H. Wang, B. Dislich, A. Colombo, U. Zeitschel et al., ADAM10 is the physiologically relevant, constitutive alpha-secretase of the amyloid precursor protein in primary neurons, EMBO J, vol.29, pp.3020-3032, 2010.

T. Laframboise, Single nucleotide polymorphism arrays: a decade of biological, computational and technological advances, Nucleic Acids Res, vol.37, pp.4181-4193, 2009.

J. Lambert, S. Heath, G. Even, D. Campion, K. Sleegers et al., , 2009.

, Genome-wide association study identifies variants at CLU aad C'' assoiated ith Alzheies disease, Nat. Geet, vol.41, pp.1094-1099

J. C. Lambert, C. A. Ibrahim-verbaas, D. Harold, A. C. Naj, R. Sims et al., Meta-analysis of 74,046 individuals idetifies e suseptiilitt loi fo Alzheies disease, Nat. Geet, vol.45, pp.1452-1458, 2013.

J. Lambert, B. Grenier-boley, D. Harold, D. Zelenika, V. Chouraki et al., , 2013.

, Genome-wide haplotype association study idetifies the F'MDA gee as a isk lous fo Alzheies disease, Mol. Pshiatt, vol.18, pp.461-470

J. Lambert, B. Grenier-boley, C. Bellenguez, F. Pasquier, D. Campion et al., PLD aad spoadi Alzheies disease isk, Natue, vol.520, p.1

M. P. Lambert, P. T. Velasco, L. Chang, K. L. Viola, S. Fernandez et al., Monoclonal antibodies that target pathological assemblies of Abeta, J. Neurochem, vol.100, pp.23-35, 2007.

E. S. Lander, L. M. Linton, B. Birren, C. Nusbaum, M. C. Zody et al., Initial sequencing and analysis of the human genome, Nature, vol.409, pp.860-921, 2001.

H. Lanoiselée, G. Nicolas, D. Wallon, A. Rovelet-lecrux, M. Lacour et al., APP, PSEN1, and PSEN2 mutations in earlyonset Alzheimer disease: A genetic screening study of familial and sporadic cases, PLoS Med, vol.14, p.1002270, 2017.

J. Laurén, 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.1128-1132, 2009.

R. M. Layer, C. Chiang, A. R. Quinlan, and I. M. Hall, LUMPY: a probabilistic framework for structural variant discovery, Genome Biol, vol.15, p.84, 2014.

L. Guennec, K. Quenez, O. Nicolas, G. Wallon, D. Rousseau et al., duplication causes prominent tau-related dementia with increased MAPT expression, Mol. Psychiatry, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01832142

M. S. Lee, Y. T. Kwon, M. Li, J. Peng, R. M. Friedlander et al., Neurotoxicity induces cleavage of p35 to p25 by calpain, Nature, vol.405, pp.360-364, 2000.

S. Lee, G. R. Abecasis, M. Boehnke, L. , and X. , Rare-variant association analysis: study designs and statistical tests, Am. J. Hum. Genet, vol.95, pp.5-23, 2014.

S. J. Van-der-lee, H. Holstege, T. H. Wong, J. Jakobsdottir, J. C. Bis et al., PLD3 variants in population studies, vol.520, pp.2-3, 2015.

M. Lek, K. J. Karczewski, E. V. Minikel, K. E. Samocha, E. Banks et al., Analysis of protein-coding genetic variation in 60,706 humans, Nature, vol.536, pp.285-291, 2016.

D. F. Levinson, J. Duan, S. Oh, K. Wang, A. R. Sanders et al., Copy number variants in schizophrenia: confirmation of five previous findings and new evidence for 3q29 microdeletions and VIPR2 duplications, Am. J. Psychiatry, vol.168, pp.302-316, 2011.

H. Li and R. Durbin, Fast and accurate short read alignment with Burrows-Wheeler transform, Bioinforma. Oxf. Engl, vol.25, pp.1754-1760, 2009.

J. Li, R. Lupat, K. C. Amarasinghe, E. R. Thompson, M. A. Doyle et al., CONTRA: copy number analysis for targeted resequencing, Bioinforma. Oxf. Engl, vol.28, pp.1307-1313, 2012.

M. Li, H. Gui, J. S. Kwan, S. Bao, and P. C. Sham, A comprehensive framework for prioritizing variants in exome sequencing studies of Mendelian diseases, Nucleic Acids Res, vol.40, p.53, 2012.

S. Li, M. Jin, T. Koeglsperger, N. E. Shepardson, G. M. Shankar et al., Soluble A? oligomers inhibit long-term potentiation through a mechanism involving excessive activation of extrasynaptic NR2B-containing NMDA receptors, J. Neurosci. Off. J. Soc. Neurosci, vol.31, pp.6627-6638, 2011.

Y. Li, C. A. Shaw, I. Sheffer, N. Sule, S. Z. Powell et al., Integrated copy number and gene expression analysis detects a C'EB assoiatio ith Alzheies disease, Taasl. Pshiatt, vol.2, p.192, 2012.

J. Lim, T. Hao, C. Shaw, A. J. Patel, G. Szabó et al., A protein-protein interaction network for human inherited ataxias and disorders of Purkinje cell degeneration, Cell, vol.125, pp.801-814, 2006.

X. Liu, X. Jian, and E. Boerwinkle, dbNSFP v2.0: a database of human non-synonymous SNVs and their functional predictions and annotations, Hum. Mutat, vol.34, pp.2393-2402, 2013.

F. Logeat, C. Bessia, C. Brou, O. Lebail, S. Jarriault et al., The Notch1 receptor is cleaved constitutively by a furin-like convertase, Proc. Natl. Acad. Sci. U. S. A, vol.95, pp.8108-8112, 1998.

E. Louwersheimer, P. E. Cohn-hokke, Y. A. Pijnenburg, M. M. Weiss, E. A. Sistermans et al., Rare Genetic Variant in "O'L Ma Iease Peetaae of Alzheies Disease i a Family with Several Generations of APOE-?4 Homozygosity, J. Alzheimers Dis. JAD, vol.56, pp.63-74, 2017.

D. G. Macarthur, S. Balasubramanian, A. Frankish, N. Huang, J. Morris et al., A systematic survey of loss-of-function variants in human protein-coding genes, Science, vol.335, pp.823-828, 2012.

J. R. Macdonald, R. Ziman, R. K. Yuen, L. Feuk, and S. W. Scherer, The Database of Genomic Variants: a curated collection of structural variation in the human genome, Nucleic Acids Res, vol.42, pp.986-992, 2014.

G. Mairet-coello, J. Courchet, S. Pieraut, V. Courchet, A. Maximov et al., The CAMKK2-AMPK kinase pathway mediates the synaptotoxic effects of A? oligomers through Tau phosphorylation, Neuron, vol.78, pp.94-108, 2013.

S. A. Malekpour, H. Pezeshk, and M. Sadeghi, MGP-HMM: Detecting genome-wide CNVs using an HMM for modeling mate pair insertion sizes and read counts, Math. Biosci, vol.279, pp.53-62, 2016.

T. A. Manolio, F. S. Collins, N. J. Cox, D. B. Goldstein, L. A. Hindorff et al., Finding the missing heritability of complex diseases, Nature, vol.461, pp.747-753, 2009.

G. Mckhann, D. Drachman, M. Folstein, R. Katzman, D. Price et al., Clinical diagosis of Alzheies disease: epot of the NINCD"-ADRDA Work Group under the auspices of Depaatet of Health aad Huaa "eies Task Foe o Alzheies Disease, Neuolog, vol.34, pp.939-944, 1984.

G. M. Mckhann, D. S. Knopman, H. Chertkow, B. T. Hyman, C. R. Jack et al., The diagosis of deetia due to Alzheies disease: recommendations from the National Institute on Aging-Alzheies Assoiatio okgoups o diagosti guidelies fo Alzheies disease, Alzheies Deet. J. Alzheies Asso, vol.7, pp.263-269, 2011.

A. Melnikov, A. Murugan, X. Zhang, T. Tesileanu, L. Wang et al., Systematic dissection and optimization of inducible enhancers in human cells using a massively parallel reporter assay, Nat. Biotechnol, vol.30, pp.271-277, 2012.

J. J. Michaelson, Y. Shi, M. Gujral, H. Zheng, D. Malhotra et al., Whole-genome sequencing in autism identifies hot spots for de novo germline mutation, Cell, vol.151, pp.1431-1442, 2012.

M. A. Mooney and B. Wilmot, Gene set analysis: A step-by-step guide, Am. J. Med. Genet. Part B Neuropsychiatr. Genet. Off. Publ. Int. Soc. Psychiatr. Genet, vol.168, pp.517-527, 2015.

G. P. Morris, I. A. Clark, and B. Vissel, Inconsistencies and controversies surrounding the aaloid hpothesis of Alzheies disease, Ata Neuopathol. Cou, vol.2, p.135, 2014.

R. Morsch, W. Simon, C. , and P. D. , Neurons may live for decades with neurofibrillary tangles, J. Neuropathol. Exp. Neurol, vol.58, pp.188-197, 1999.

A. C. Naj, G. Jun, G. W. Beecham, L. Wang, B. N. Vardarajan et al., Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-oset Alzheies disease, Nat. Geet, vol.43, pp.436-441, 2011.

A. C. Naj and G. D. , aad Alzheies Disease Geetis Cosotiu ADGC. Geoi aaiaats, gees, aad pathaas of Alzheies disease: A oeiew, Am. J. Med. Genet. Part B Neuropsychiatr. Genet. Off. Publ. Int. Soc. Psychiatr. Genet, vol.174, pp.5-26

J. Nam, N. K. Kim, S. C. Kim, J. Joung, R. Xi et al., Evaluation of somatic copy number estimation tools for whole-exome sequencing data, Brief. Bioinform, vol.17, pp.185-192, 2016.

P. C. Ng and S. Henikoff, Accounting for human polymorphisms predicted to affect protein function, Genome Res, vol.12, pp.436-446, 2002.

P. C. Ng and S. Henikoff, SIFT: Predicting amino acid changes that affect protein function, Nucleic Acids Res, vol.31, pp.3812-3814, 2003.

S. B. Ng, E. H. Turner, P. D. Robertson, S. D. Flygare, A. W. Bigham et al., Targeted capture and massively parallel sequencing of 12 human exomes, Nature, vol.461, pp.272-276, 2009.

G. Nicolas, D. Wallon, C. Charbonnier, O. Quenez, S. Rousseau et al., Screening of dementia genes by whole-exome sequencing in early-onset Alzheimer disease: input and lessons, Eur. J. Hum. Genet. EJHG, vol.24, pp.710-716, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01431285

G. Nicolas, C. Charbonnier, D. Wallon, O. Quenez, C. Bellenguez et al., SORL1 rare variants: a major risk factor for familial early-oset Alzheies disease, Mol. Psychiatry, vol.21, pp.831-836, 2016.

G. Nicolas, D. Wallon, C. Goupil, A. Richard, C. Pottier et al., Mutatio i the uttaaslated egio of APP as a geeti determinant of cerebral amyloid angiopathy, Eur. J. Hum. Genet. EJHG, vol.24, pp.92-98

C. Nilsberth, A. Westlind-danielsson, C. B. Eckman, M. M. Condron, K. Axelman et al., The Ati APP utatio EG auses Alzheies disease ehaaed Aeta potofiil foatio, Nat. Neuosi, vol.4, pp.887-893

R. M. Nisbet, J. Polanco, L. M. Ittner, G. , and J. , Tau aggregation and its interplay with amyloid-?, Acta Neuropathol. (Berl.), vol.129, pp.207-220, 2015.

G. Novarino, A. G. Fenstermaker, M. S. Zaki, M. Hofree, J. L. Silhavy et al., Exome sequencing links corticospinal motor neuron disease to common neurodegenerative disorders, Science, vol.343, pp.506-511, 2014.

M. C. Oh, V. A. Derkach, E. S. Guire, and T. R. Soderling, Extrasynaptic membrane trafficking regulated by GluR1 serine 845 phosphorylation primes AMPA receptors for long-term potentiation, J. Biol. Chem, vol.281, pp.752-758, 2006.

F. Oyama, N. Sawamura, K. Kobayashi, M. Morishima-kawashima, T. Kuramochi et al., Mutant presenilin 2 transgenic mouse: effect on an age-dependent increase of amyloid beta-protein 42 in the brain, J. Neurochem, vol.71, pp.313-322, 1998.

R. P. Patwardhan, J. B. Hiatt, D. M. Witten, M. J. Kim, R. P. Smith et al., Massively parallel functional dissection of mammalian enhancers in vivo, Nat. Biotechnol, vol.30, pp.265-270, 2012.

J. Perez-tur, S. Froelich, G. Prihar, R. Crook, M. Baker et al., A utatio i Alzheies disease desttoig a splie aaepto site i the presenilin-1 gene, Neuroreport, vol.7, pp.297-301

L. Piccio, Y. Deming, J. L. Del-Águila, L. Ghezzi, D. M. Holtzman et al., Cerebrospinal fluid soluble TREM2 is higher in Alzheimer disease and associated with mutation status, Acta Neuropathol. (Berl.), vol.131, pp.925-933, 2016.

D. Pinkel, A. , and D. G. , Comparative genomic hybridization, Annu. Rev. Genomics Hum. Genet, vol.6, pp.331-354, 2005.

V. Plagnol, J. Curtis, M. Epstein, K. Y. Mok, E. Stebbings et al., A robust model for read count data in exome sequencing experiments and implications for copy number variant calling, Bioinforma. Oxf. Engl, vol.28, pp.2747-2754, 2012.

A. J. Pocklington, E. Rees, J. T. Walters, J. Han, D. H. Kavanagh et al., Novel Findings from CNVs Implicate Inhibitory and Excitatory Signaling Complexes in Schizophrenia, Neuron, vol.86, pp.1203-1214, 2015.

C. Pottier, D. Wallon, A. R. Lecrux, D. Maltete, S. Bombois et al., Amyloid-? potei peuso gee epessio i alzheies disease aad other conditions, J. Alzheimers Dis. JAD, vol.28, pp.561-566, 2012.

C. Pottier, D. Hannequin, S. Coutant, A. Rovelet-lecrux, D. Wallon et al., High frequency of potentially pathogenic SORL1 mutations in autosomal dominant early-onset Alzheimer disease, Mol. Psychiatry, vol.17, pp.875-879, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00965204

M. Rapoport, H. N. Dawson, L. I. Binder, M. P. Vitek, and A. Ferreira, Tau is essential to betaamyloid-induced neurotoxicity, Proc. Natl. Acad. Sci. U. S. A, vol.99, pp.6364-6369, 2002.

R. Redon, S. Ishikawa, K. R. Fitch, L. Feuk, G. H. Perry et al., Global variation in copy number in the human genome, Nature, vol.444, pp.444-454, 2006.

C. Reitz, The role of the retromer complex in aging-related neurodegeneration: a molecular and genomic review, Mol. Genet. Genomics MGG, vol.290, pp.413-427, 2015.

S. Richards, N. Aziz, S. Bale, D. Bick, S. Das et al., Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology, Genet. Med. Off. J. Am. Coll. Med. Genet, vol.17, pp.405-424, 2015.

S. L. Rosenthal, M. N. Bamne, X. Wang, S. Berman, B. E. Snitz et al., More evidence for association of a rare TREM2 mutation ''H ith Alzheies disease isk, Neurobiol. Aging, vol.36, pp.2443-2464, 2015.

A. Rovelet-lecrux, D. Hannequin, G. Raux, N. Le-meur, A. Laquerrière et al., APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy, Nat. Genet, vol.38, pp.24-26, 2006.

A. Rovelet-lecrux, S. Legallic, D. Wallon, J. Flaman, O. Martinaud et al., A genome-wide study reveals rare CNVs exclusive to extreme phenotypes of Alzheimer disease, Eur. J. Hum. Genet. EJHG, vol.20, pp.613-617, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00965208

A. Rovelet-lecrux, C. Charbonnier, D. Wallon, G. Nicolas, M. N. Seaman et al., De novo deleterious genetic variations target a biological network centered on A? peptide in early-onset Alzheimer disease, Mol. Psychiatry, vol.20, pp.1046-1056, 2015.

D. M. Ruderfer, T. Hamamsy, M. Lek, K. J. Karczewski, D. Kavanagh et al., Patterns of genic intolerance of rare copy number variation in 59,898 human exomes, Nat. Genet, vol.48, pp.1107-1111, 2016.

T. Saito, T. Suemoto, N. Brouwers, K. Sleegers, S. Funamoto et al., Potent amyloidogenicity and pathogenicity of A?43, Nat. Neurosci, vol.14, pp.1023-1032, 2011.

P. S. Samarakoon, H. S. Sorte, A. Stray-pedersen, O. K. Rødningen, T. Rognes et al., cnvScan: a CNV screening and annotation tool to improve the clinical utility of computational CNV prediction from exome sequencing data, BMC Genomics, vol.17, p.51, 2016.

F. Sanger and A. R. Coulson, A rapid method for determining sequences in DNA by primed synthesis with DNA polymerase, J. Mol. Biol, vol.94, pp.441-448, 1975.

F. Sanger, S. Nicklen, and A. R. Coulson, DNA sequencing with chain-terminating inhibitors, Proc. Natl. Acad. Sci. U. S. A, vol.74, pp.5463-5467, 1977.

R. Sannerud, C. Esselens, P. Ejsmont, R. Mattera, L. Rochin et al., Restricted Location of PSEN2/?-Secretase Determines Substrate Specificity and Generates an Intracellular A? Pool, Cell, vol.166, pp.193-208, 2016.

S. Sato, K. Kamino, T. Miki, A. Doi, K. Ii et al., Splicing mutation of presenilin-1 gene for early-oset faailial Alzheies disease, Hu. Mutat. Suppl, vol.1, pp.91-94, 1998.

K. Satoh, S. Abe-dohmae, S. Yokoyama, P. St-george-hyslop, and P. E. Fraser, ATP-binding cassette transporter A7 (ABCA7) loss of function alters Alzheimer amyloid processing, J. Biol. Chem, vol.290, pp.24152-24165, 2015.

N. Sawamura, M. Morishima-kawashima, H. Waki, K. Kobayashi, T. Kuramochi et al., Mutant presenilin 2 transgenic mice. A large increase in the levels of Abeta 42 is presumably associated with the low density membrane domain that contains decreased levels of glycerophospholipids and sphingomyelin, J. Biol. Chem, vol.275, pp.27901-27908, 2000.

G. D. Schellenberg, S. S. Deeb, M. Boehnke, E. M. Bryant, G. M. Martin et al., Association of an apolipoprotein CII allele with familial dementia of the Alzheimer type, J. Neurogenet, vol.4, pp.97-108, 1987.

D. Scheuner, C. Eckman, M. Jensen, X. Song, M. Citron et al., Secreted amyloid beta-protein similar to that in the senile plaques of Alzheies disease is ieased i io the presenilin 1 and 2 and APP mutations linked to familial Alzheies disease, Nat. Med, vol.2, pp.864-870, 1996.

A. Schlattl, S. Anders, S. M. Waszak, W. Huber, and J. O. Korbel, Relating CNVs to transcriptome data at fine resolution: assessment of the effect of variant size, type, and overlap with functional regions, Genome Res, vol.21, 2004.

E. C. Schulte, A. Kurz, P. Alexopoulos, H. Hampel, A. Peters et al., Excess of rare coding variants in PLD3 in late-but not early-onset Alzheies disease. Hu. Geoe Vaa, vol.2, 2015.

J. M. Schwarz, C. Rödelsperger, M. Schuelke, and D. Seelow, MutationTaster evaluates disease-causing potential of sequence alterations, Nat. Methods, vol.7, pp.575-576, 2010.

J. M. Schwarz, D. N. Cooper, M. Schuelke, and D. Seelow, MutationTaster2: mutation prediction for the deep-sequencing age, Nat. Methods, vol.11, pp.361-362, 2014.

M. N. Seaman, The retromer complex-endosomal protein recycling and beyond, J. Cell Sci, vol.125, pp.4693-4702, 2012.

J. Sebat, B. Lakshmi, J. Troge, J. Alexander, J. Young et al., Large-scale copy number polymorphism in the human genome, Science, vol.305, pp.525-528, 2004.

S. Seshadri, A. L. Fitzpatrick, M. A. Ikram, A. L. Destefano, V. Gudnason et al., , 2010.

, Genome-wide analysis of genetic loci associated with Alzheimer disease, JAMA, vol.303, pp.1832-1840

G. M. Shankar, B. L. Bloodgood, M. Townsend, D. M. Walsh, D. J. Selkoe et al., Natural oligomers of the Alzheimer amyloid-beta protein induce reversible synapse loss by modulating an NMDA-type glutamate receptor-dependent signaling pathway, J. Neurosci. Off. J. Soc. Neurosci, vol.27, pp.2866-2875, 2007.

G. M. Shankar, S. Li, T. H. Mehta, A. Garcia-munoz, N. E. Shepardson et al., Amyloid-beta protein dimers isolated directly from Alzheies ais ipai sapti plastiitt aad eo, Nat. Med, vol.14, pp.837-842, 2008.

C. Shaw-smith, A. M. Pittman, L. Willatt, H. Martin, L. Rickman et al., Microdeletion encompassing MAPT at chromosome 17q21.3 is associated with developmental delay and learning disability, Proc. Natl. Acad. Sci. U. S. A, vol.38, pp.403-409, 2006.

S. S. Sisodia, E. H. Koo, K. Beyreuther, A. Unterbeck, P. et al., Evidence that betaaaloid potei i Alzheies disease is ot deied oal poessig, vol.248, pp.492-495, 1990.

M. J. Smith, J. B. Kwok, C. A. Mclean, J. J. Kril, G. A. Broe et al., Vaaiaale pheottpe of Alzheies disease ith spasti paraparesis, Ann. Neurol, vol.49, pp.125-129

E. M. Snyder, Y. Nong, C. G. Almeida, S. Paul, T. Moran et al., Regulation of NMDA receptor trafficking by amyloid-beta, Nat. Neurosci, vol.8, pp.1051-1058, 2005.

Y. Sottejeau, A. Bretteville, F. Cantrelle, N. Malmanche, F. Demiaute et al., Tau phosphorylation regulates the interaction etee BINs "H doai aad Taus polie-rich domain, Acta Neuropathol. Commun, vol.3, p.58, 2015.

T. L. Spires-jones, A. De-calignon, T. Matsui, C. Zehr, R. Pitstick et al., In vivo imaging reveals dissociation between caspase activation and acute neuronal death in tangle-bearing neurons, J. Neurosci. Off. J. Soc. Neurosci, vol.28, pp.862-867, 2008.

S. George-hyslop, P. H. Tanzi, R. E. Polinsky, R. J. Haines, J. L. Nee et al., The genetic defect causing familial Alzheies disease maps on chromosome 21, Science, vol.235, pp.885-890, 1987.

S. Steinberg, H. Stefansson, T. Jonsson, H. Johannsdottir, A. Ingason et al., Loss-of-function variants in ABCA ofe isk of Alzheies disease, Nat. Geet, vol.47, pp.445-447, 2015.

M. Suárez-calvet, G. Kleinberger, M. Á. Araque-caballero, M. Brendel, A. Rominger et al., sTREM2 cerebrospinal fluid levels are a potential biomarker for microglia activity in early-stage Alzheies disease aad assoiate ith neuronal injury markers, EMBO Mol. Med, vol.8, pp.466-476, 2016.

S. Swaminathan, M. J. Huentelman, J. J. Corneveaux, A. J. Myers, K. M. Faber et al., Analysis of op ue aaiatio i Alzheies disease i a cohort of clinically characterized and neuropathologically verified individuals, PloS One, vol.7, p.50640, 2012.

S. Swaminathan, L. Shen, S. Kim, M. Inlow, J. D. West et al., Aalsis of op ue aaiatio i Alzheies disease: the NIALOAD/ NC'AD Fail, Alzheies Disease Neuoiagig Iitiatie, vol.9, pp.801-814

M. Szaruga, S. Veugelen, M. Benurwar, S. Lismont, D. Sepulveda-falla et al., Qualitative changes in human ?-secretase underlie familial Alzheies disease, Mol. Psychiatry, vol.212, pp.762-773, 2003.
DOI : 10.1084/jem.20150892

URL : http://jem.rupress.org/content/jem/212/12/2003.full.pdf

K. Szigeti, D. Lal, Y. Li, R. S. Doody, K. Wilhelmsen et al., Genome-wide scan for copy number variation association ith age at oset of Alzheies disease, J. Alzheies Dis. JAD, vol.33, pp.517-523, 2013.

M. Takami, Y. Nagashima, Y. Sano, S. Ishihara, M. Morishima-kawashima et al., gamma-Secretase: successive tripeptide and tetrapeptide release from the transmembrane domain of beta-carboxyl terminal fragment, J. Neurosci. Off. J. Soc. Neurosci, vol.29, pp.13042-13052, 2009.

R. Tan, Y. Wang, S. E. Kleinstein, Y. Liu, X. Zhu et al., An evaluation of copy number variation detection tools from whole-exome sequencing data, Hum. Mutat, vol.35, pp.899-907, 2014.
DOI : 10.1002/humu.22537

T. Tapiola, I. Alafuzoff, S. Herukka, L. Parkkinen, P. Hartikainen et al., Cerebrospinal fluid {beta}-amyloid 42 and tau proteins as biomarkers of Alzheimer-type pathologic changes in the brain, Arch. Neurol, vol.66, pp.382-389, 2009.

J. A. Teesse, A. W. Bighaa, T. D. Ocoo, W. Fu, E. E. Kenny et al., Evolution and functional impact of rare coding variation from deep sequencing of human exomes, Science, vol.337, pp.64-69, 2012.

D. R. Thal, U. Rüb, M. Orantes, and H. Braak, Phases of A beta-deposition in the human brain and its relevance for the development of AD, Neurology, vol.58, pp.1791-1800, 2002.

G. Thinakaran, D. R. Borchelt, M. K. Lee, H. H. Slunt, L. Spitzer et al., Endoproteolysis of presenilin 1 and accumulation of processed derivatives in vivo, Neuron, vol.17, pp.181-190, 1996.

J. Thusberg, A. Olatubosun, and M. Vihinen, Performance of mutation pathogenicity prediction methods on missense variants, Hum. Mutat, vol.32, pp.358-368, 2011.
DOI : 10.1002/humu.21445

URL : http://onlinelibrary.wiley.com/doi/10.1002/humu.21445/pdf

G. Tosto, H. Fu, B. N. Vardarajan, J. H. Lee, R. Cheng et al., F-box/LRR-repeat protein 7 is genetically assoiated ith Alzheies disease, A. Clin. Transl. Neurol, vol.2, pp.810-820, 2015.
DOI : 10.1002/acn3.223

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1002/acn3.223

K. L. Tucker, M. Meyer, B. , and Y. A. , Neurotrophins are required for nerve growth during development, Nat. Neurosci, vol.4, pp.29-37, 2001.
DOI : 10.1038/82868

F. Ubelmann, T. Burrinha, L. Salavessa, R. Gomes, C. Ferreira et al., Bin1 and CD2AP polarise the endocytic generation of beta-amyloid, EMBO Rep, vol.18, pp.102-122, 2017.
DOI : 10.15252/embr.201642738

URL : http://embor.embopress.org/content/18/1/102.full.pdf

. Uk10k-consortium, K. Walter, J. L. Min, J. Huang, L. Crooks et al., The UK10K project identifies rare variants in health and disease, Nature, vol.526, pp.82-90, 2015.

J. D. Ulrich, M. B. Finn, Y. Wang, A. Shen, T. E. Mahan et al., Altered microglial response to A? plaques in APPPS1-21 mice heterozygous for TREM2, Mol. Neurodegener, vol.9, p.20, 2014.

C. Van-cauwenberghe, C. Van-broeckhoven, and K. Sleegers, The genetic landscape of Alzheimer disease: clinical implications and perspectives, Genet. Med. Off. J. Am. Coll. Med. Genet, vol.18, pp.421-430, 2016.

G. A. Van-der-auwera, M. O. Carneiro, C. Hartl, R. Poplin, G. Del-angel et al., From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline, Curr. Protoc. Bioinforma, vol.43, pp.11-21, 2013.

K. Van-schil, M. Karlstetter, A. Aslanidis, K. Dannhausen, M. Azam et al., Autosomal recessive retinitis pigmentosa with homozygous rhodopsin mutation E150K and non-coding cis-regulatory variants in CRX-binding regions of SAMD7, Sci. Rep, vol.6, p.21307, 2016.

B. N. Vardarajan, Y. Zhang, J. H. Lee, R. Cheng, C. Bohm et al., Coding mutations in SORL1 and Alzheimer disease, Ann. Neurol, vol.77, pp.215-227, 2015.

V. Vasiliou, K. Vasiliou, and D. W. Nebert, Human ATP-binding cassette (ABC) transporter family, Hum. Genomics, vol.3, pp.281-290, 2009.
DOI : 10.1186/1479-7364-3-3-281

URL : https://humgenomics.biomedcentral.com/track/pdf/10.1186/1479-7364-3-3-281

R. Vassar, BACE1: the beta-secretase enze i Alzheies disease, J. Mol. Neuosi. MN, vol.23, pp.105-114, 2004.
DOI : 10.1186/1750-1326-7-s1-l3

URL : https://molecularneurodegeneration.biomedcentral.com/track/pdf/10.1186/1750-1326-7-S1-L3?site=molecularneurodegeneration.biomedcentral.com

R. Vassar, B. D. Bennett, S. Babu-khan, S. Kahn, E. A. Mendiaz et al., Beta-seetase leaaage of Alzheies aaloid pecursor protein by the transmembrane aspartic protease BACE, Science, vol.286, pp.735-741, 1999.

J. A. Veltman and H. G. Brunner, De novo mutations in human genetic disease, Nat. Rev. Genet, vol.13, pp.565-575, 2012.
DOI : 10.1038/nrg3241

J. C. Venter, M. D. Adams, E. W. Myers, P. W. Li, R. J. Mural et al., The sequence of the human genome, Science, vol.291, pp.1304-1351, 2001.
URL : https://hal.archives-ouvertes.fr/hal-00465088

K. S. Vetrivel and G. Thinakaran, Amyloidogenic processing of beta-amyloid precursor protein in intracellular compartments, Neurology, vol.66, pp.69-73, 2006.

S. Veugelen, T. Saito, T. C. Saido, L. Chávez-gutiérrez, D. Strooper et al., Familial Alzheies Disease Mutatios i Peseili Geeate Aloidogenic A? Peptide Seeds, Neuron, vol.90, pp.410-416, 2016.

M. Vidal, M. E. Cusick, and A. Barabási, Interactome networks and human disease, Cell, vol.144, pp.986-998, 2011.
DOI : 10.1016/j.cell.2011.02.016

URL : https://doi.org/10.1016/j.cell.2011.02.016

C. M. Vockley, C. Guo, W. H. Majoros, M. Nodzenski, D. M. Scholtens et al., Massively parallel quantification of the regulatory effects of noncoding genetic variation in a human cohort, Genome Res, vol.25, pp.1206-1214, 2015.

V. Vogelsberg-ragaglia, J. Bruce, C. Richter-landsberg, B. Zhang, M. Hong et al., Distinct FTDP-17 missense mutations in tau produce tau aggregates and other pathological phenotypes in transfected CHO cells, Mol. Biol. Cell, vol.11, pp.4093-4104, 2000.
DOI : 10.1091/mbc.11.12.4093

URL : http://www.molbiolcell.org/content/11/12/4093.full.pdf

D. Wallon, S. Rousseau, A. Rovelet-lecrux, M. Quillard-muraine, L. Guyant-maréchal et al., The French series of autosomal doiaat eaal oset Alzheies disease ases: utatio spettu aad eeospial fluid biomarkers, J. Alzheimers Dis. JAD, vol.30, pp.847-856, 2012.

D. M. Walsh and D. J. Selkoe, A beta oligomers-a decade of discovery, J. Neurochem, vol.101, pp.1172-1184, 2007.
DOI : 10.1111/j.1471-4159.2006.04426.x

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1471-4159.2006.04426.x/pdf

Y. Wang and E. Mandelkow, Tau in physiology and pathology, Nat. Rev. Neurosci, vol.17, pp.5-21, 2016.
DOI : 10.1038/nrn.2015.1

B. Wang, W. Yang, W. Wen, J. Sun, B. Su et al., , 2010.

, Gamma-secretase gene mutations in familial acne inversa, Science, vol.330, p.1065

H. Y. Wang, D. H. Lee, C. B. Davis, and R. P. Shank, Amyloid peptide Abeta(1-42) binds selectively and with picomolar affinity to alpha7 nicotinic acetylcholine receptors, J. Neurochem, vol.75, pp.1155-1161, 2000.

Q. Wang, Q. Lu, and H. Zhao, A review of study designs and statistical methods for genomic epidemiology studies using next generation sequencing, Front. Genet, vol.6, p.149, 2015.

Y. Wang, M. Cella, K. Mallinson, J. D. Ulrich, K. L. Young et al., TREM2 lipid sensing sustains the microglial espose i aa Alzheies disease odel, Cell, vol.160, pp.1061-1071, 2015.

Y. Wang, T. K. Ulland, J. D. Ulrich, W. Song, J. A. Tzaferis et al., TREM2-mediated early microglial response limits diffusion and toxicity of amyloid plaques, J. Exp. Med, vol.213, pp.667-675, 2016.

M. D. Weingarten, A. H. Lockwood, S. Y. Hwo, and M. W. Kirschner, A protein factor essential for microtubule assembly, Proc. Natl. Acad. Sci. U. S. A, vol.72, pp.1858-1862, 1975.

P. J. Whitehouse, D. L. Price, A. W. Clark, J. T. Coyle, and M. R. Delong, Alzheimer disease: evidence for selective loss of cholinergic neurons in the nucleus basalis, Ann. Neurol, vol.10, pp.122-126, 1981.

M. S. Wolfe, W. Xia, B. L. Ostaszewski, T. S. Diehl, W. T. Kimberly et al., Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and gammasecretase activity, Nature, vol.398, pp.513-517, 1999.

D. Xia, H. Watanabe, B. Wu, S. H. Lee, Y. Li et al., Presenilin-1 knockin mice reveal loss-of-futio ehaais fo failial Alzheies disease, Neuron, vol.85, pp.967-981, 2015.

D. Xia, R. J. Kelleher, and J. Shen, Loss of A?43 Production Caused by Presenilin-1 Mutations in the Knockin Mouse Brain, Neuron, vol.90, pp.417-422, 2016.

J. Yu, M. A. Vodyanik, K. Smuga-otto, J. Antosiewicz-bourget, J. L. Frane et al., Induced pluripotent stem cell lines derived from human somatic cells, Science, vol.318, pp.1917-1920, 2007.

J. Yu, L. Taa, and J. Haad,

E. Apolipopotei, Alzheies disease: aa update, Au. 'e. Neurosci, vol.37, pp.79-100

W. Yu, J. Polepalli, D. Wagh, J. Rajadas, R. Malenka et al., A critical role for the PAR1/MARK-tau axis in mediating the toxic effects of A? on synapses and dendritic spines, Hum. Mol. Genet, vol.21, pp.1384-1390, 2012.

P. Yuan, C. Condello, C. D. Keene, Y. Wang, T. D. Bird et al., TREM2 Haplodeficiency in Mice and Humans Impairs the Microglia Barrier Function Leading to Decreased Amyloid Compaction and Severe Axonal Dystrophy, Neuron, vol.92, pp.252-264, 2016.

Z. Zappala, M. , and S. B. , Non-Coding Loss-of-Function Variation in Human Genomes, Hum. Hered, vol.81, pp.78-87, 2016.

M. Zarrei, J. R. Macdonald, D. Merico, and S. W. Scherer, A copy number variation map of the human genome, Nat. Rev. Genet, vol.16, pp.172-183, 2015.

D. Zhang, Y. Fan, D. Wang, R. Bi, C. Zhang et al., PLD i Alzheies Disease: a Modest Effect as Revealed by Updated Association and Expression Analyses, Mol. Neurobiol, vol.53, pp.4034-4045

W. Zhang, B. Jiao, M. Zhou, T. Zhou, and L. He,

, Modelig Alzheies Disease ith Induced Pluripotent Stem Cells: Current Challenges and Future Concerns, Stem Cells Int, p.7828049, 2016.

M. Zhao, Q. Wang, Q. Wang, P. Jia, and Z. Zhao, Computational tools for copy number variation (CNV) detection using next-generation sequencing data: features and perspectives, BMC Bioinformatics, vol.14, p.1, 2013.

L. Zhong, X. Chen, T. Wang, Z. Wang, C. Liao et al., Soluble TREM2 induces inflammatory responses and enhances microglial survival, J. Exp. Med, vol.214, pp.597-607, 2017.

X. Zhu, J. Yu, T. Jiaag, P. Waag, L. Cao et al., C' i Alzheies disease, Mol. Neurobiol, vol.51, pp.753-765

M. Zollino, D. Orteschi, M. Murdolo, S. Lattante, D. Battaglia et al., Mutations in KANSL1 cause the 17q21.31 microdeletion syndrome phenotype, Nat. Genet, vol.44, pp.636-638, 2012.