, Centre National de la Recherche Scientifique (CNRS) UMR 5310, Institut Neuromyogène, Synaptopathies and Autoantibodies, Neuromyogene Institut, F-69000 Lyon, France; 2) Université Claude Bernard Lyon 1, F-69000 Lyon, France ; 3) INSERM U1052, CNRS UMR5286, Cancer Cell Plasticity, Cancer Research Centre of Lyon, F-69000 Lyon, France ; 4) Centre de Pathologie et de Neuropathologie Est, Hospices Civils de Lyon F-69000 Lyon, France; 5) GIN, Inserm U1216, INSERM Unité, vol.38000, issue.6

S. Alfonso, H. W. Kessels, C. C. Banos, T. R. Chan, E. T. Lin et al., Synapto-depressive effects of amyloid beta require PICK1, Eur J Neurosci, vol.39, pp.1225-1258, 2014.

C. G. Almeida, D. Tampellini, R. H. Takahashi, P. Greengard, M. T. Lin et al., Beta-amyloid accumulation in APP mutant neurons reduces PSD-95 and GluR1 in synapses, Neurobiol Dis, vol.20, pp.187-98, 2005.

M. F. Bear and R. C. Malenka, Synaptic plasticity: LTP and LTD, Curr Opin Neurobiol, vol.4, pp.389-99, 1994.

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.1291-300, 2000.

J. Bourne and K. M. Harris, Do thin spines learn to be mushroom spines that remember?, Curr Opin Neurobiol, vol.17, pp.381-387, 2007.

H. Braak and E. Braak, Demonstration of amyloid deposits and neurofibrillary changes in whole brain sections, Brain Pathol, vol.1, pp.213-219, 1991.

L. Cao, X. Jiao, D. S. Zuzga, Y. Liu, D. M. Fong et al., VEGF links hippocampal activity with neurogenesis, learning and memory, Nat Genet, vol.36, pp.827-862, 2004.

A. Citri and R. C. Malenka, Synaptic plasticity: multiple forms, functions, and mechanisms, Neuropsychopharmacology, vol.33, pp.18-41, 2008.

M. D'amelio, V. Cavallucci, S. Middei, C. Marchetti, S. Pacioni et al., Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer's disease, Nat Neurosci, vol.14, pp.69-76, 2011.

D. Rossi, P. Harde, E. Dupuis, J. P. Martin, L. Chounlamountri et al., A critical role for VEGF and VEGFR2 in NMDA receptor synaptic function and fear-related behavior, Mol Psychiatry, vol.21, pp.1768-1780, 2016.

M. M. Dorostkar, C. Zou, L. Blazquez-llorca, and J. Herms, Analyzing dendritic spine pathology in Alzheimer's disease: problems and opportunities, Acta Neuropathol, vol.130, pp.1-19, 2015.

B. Dubois, H. H. Feldman, C. Jacova, H. Hampel, J. L. Molinuevo et al., Advancing research diagnostic criteria for Alzheimer's disease: the IWG-2 criteria, Lancet Neurol, vol.13, pp.614-643, 2014.

M. D. Ehlers, Reinsertion or degradation of AMPA receptors determined by activity-dependent endocytic sorting, Neuron, vol.28, pp.511-536, 2000.

F. El-gaamouch, A. Buisson, O. Moustié, M. Lemieux, S. Labrecque et al., Interaction between ?CaMKII and GluN2B controls ERK-dependent plasticity, J Neurosci, vol.32, pp.10767-79, 2012.

J. A. Esteban, S. H. Shi, C. Wilson, M. Nuriya, R. L. Huganir et al., PKA phosphorylation of AMPA receptor subunits controls synaptic trafficking underlying plasticity, Nat Neurosci, vol.6, pp.136-179, 2003.

N. Ferrara, H. P. Gerber, and J. Lecouter, The biology of VEGF and its receptors, Nat Med, vol.9, pp.669-76, 2003.

M. L. Frandemiche, D. Seranno, S. Rush, T. Borel, E. Elie et al., Activity-dependent tau protein translocation to excitatory synapse is disrupted by exposure to amyloid-beta oligomers, J Neurosci, vol.34, pp.6084-97, 2014.

K. O. Garcia, F. L. Ornellas, P. K. Martin, C. L. Patti, L. E. Mello et al., Therapeutic effects of the transplantation of VEGF overexpressing bone marrow mesenchymal stem cells in the hippocampus of murine model of Alzheimer's disease, Front Aging Neurosci, vol.6, p.30, 2014.

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

E. Herrán, R. Pérez-gonzález, M. Igartua, J. L. Pedraz, E. Carro et al., VEGF-releasing biodegradable nanospheres administered by craniotomy: a novel therapeutic approach in the APP/Ps1 mouse model of Alzheimer's disease, J Control Release, vol.170, pp.111-120, 2013.

T. J. Hohman, S. P. Bell, and A. L. Jefferson,

, Alzheimer's Disease Neuroimaging Initiative (2015) The role of vascular endothelial growth factor in neurodegeneration and cognitive decline: exploring interactions with biomarkers of Alzheimer disease, JAMA Neurol, vol.72, pp.520-529

K. A. Houck, D. W. Leung, A. M. Rowland, J. Winer, and N. Ferrara, Dual regulation of vascular endothelial growth factor bioavailability by genetic and proteolytic mechanisms, J Biol Chem, vol.267, pp.26031-26038, 1992.

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.831-874, 2006.

H. H. Jarosz-griffiths, E. Noble, J. V. Rushworth, and N. M. Hooper, Amyloid-? Receptors: The Good, the Bad, and the Prion Protein, J Biol Chem, vol.291, pp.3174-83, 2016.

H. Jia, S. Jezequel, M. Löhr, S. Shaikh, D. Davis et al., Peptides encoded by exon 6 of VEGF inhibit endothelial cell biological responses and angiogenesis induced by VEGF, Biochem Biophys Res Commun, vol.283, pp.164-73, 2001.

H. Kasai, T. Hayama, M. Ishikawa, S. Watanabe, S. Yagishita et al., Learning rules and persistence of dendritic spines, Eur J Neurosci, vol.32, pp.241-250, 2010.

B. W. Kim, M. Choi, Y. S. Kim, H. Park, H. R. Lee et al., Vascular endothelial growth factor (VEGF) signaling regulates hippocampal neurons by elevation of intracellular calcium and activation of calcium/calmodulin protein kinase II and mammalian target of rapamycin, Cell Signal, vol.20, pp.714-739, 2008.

S. Knafo, L. Alonso-nanclares, J. Gonzalez-soriano, P. Merino-serrais, I. Fernaud-espinosa et al., Widespread changes in dendritic spines in a model of Alzheimer's disease, Cereb Cortex, vol.19, pp.586-92, 2009.

R. M. Koffie, T. Hashimoto, H. C. Tai, K. R. Kay, A. Serrano-pozo et al., Apolipoprotein E4 effects in Alzheimer's disease are mediated by synaptotoxic oligomeric amyloid-?, Brain, vol.135, pp.2155-68, 2012.

C. C. Liu, N. Zhao, Y. Yamaguchi, J. R. Cirrito, T. Kanekiyo et al., Neuronal heparan sulfates promote amyloid pathology by modulating brain amyloid-? clearance and aggregation in Alzheimer's disease, Sci Transl Med, vol.8, pp.332-376, 2016.

L. F. Lue, Y. M. Kuo, A. E. Roher, L. Brachova, Y. Shen et al., Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer's disease, Am J Pathol, vol.155, pp.853-62, 1999.

R. Malinow, AMPA receptor trafficking and long-term potentiation, Philos Trans R Soc Lond B Biol Sci, vol.358, pp.707-721, 2003.

M. Matsuzaki, N. Honkura, G. C. Ellis-davies, and H. Kasai, Structural basis of long-term potentiation in single dendritic spines, Nature, vol.429, pp.761-767, 2004.

C. A. Mclean, R. A. Cherny, F. W. Fraser, S. J. Fuller, M. J. Smith et al., Soluble pool of Abeta amyloid as a determinant of severity of neurodegeneration in Alzheimer's disease, Ann Neurol, vol.46, pp.860-866, 1999.

Z. Mi, E. E. Abrahamson, A. Y. Ryu, K. N. Fish, R. A. Sweet et al., Loss of precuneus dendritic spines immunopositive for spinophilin is related to cognitive impairment in early Alzheimer's disease, Neurobiol Aging, vol.55, pp.159-166, 2017.

A. J. Miñano-molina, J. España, E. Martín, B. Barneda-zahonero, R. Fadó et al., Soluble oligomers of amyloid-? peptide disrupt membrane trafficking of ?-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor contributing to early synapse dysfunction, J Biol Chem, vol.286, pp.27311-27332, 2011.

N. Mukerjee, K. M. Mcginnis, Y. H. Park, M. E. Gnegy, and K. K. Wang, Caspase-mediated proteolytic activation of calcineurin in thapsigargin-mediated apoptosis in SH-SY5Y neuroblastoma cells, Arch Biochem Biophys, vol.379, pp.337-380, 2000.

P. J. Nestor, P. Scheltens, and J. R. Hodges, Advances in the early detection of Alzheimer's disease, Nat Med, vol.10, pp.34-41, 2004.

P. Opazo, S. Labrecque, C. M. Tigaret, A. Frouin, P. W. Wiseman et al., CaMKII triggers the diffusional trapping of surface AMPARs through phosphorylation of stargazin, Neuron, vol.67, pp.239-52, 2010.

D. Putcha, M. Brickhouse, K. O'keefe, C. Sullivan, D. Rentz et al., Hippocampal hyperactivation associated with cortical thinning in Alzheimer's disease signature regions in non-demented elderly adults, J Neurosci, vol.31, pp.17680-17688, 2011.

R. Radde, T. Bolmont, S. A. Kaeser, J. Coomaraswamy, D. Lindau et al., Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology, EMBO Rep, vol.7, pp.940-946, 2006.

P. Religa, R. Cao, D. Religa, Y. Xue, N. Bogdanovic et al., VEGF significantly restores impaired memory behavior in Alzheimer's mice by improvement of vascular survival, Sci Rep, vol.3, p.2053, 2013.

M. Renner, P. N. Lacor, P. T. Velasco, J. Xu, A. Contractor et al., Deleterious effects of amyloid beta oligomers acting as an extracellular scaffold for mGluR5, Neuron, vol.66, pp.739-54, 2010.

K. W. Roche, R. J. O'brien, A. L. Mammen, J. Bernhardt, and R. L. Huganir, Characterization of multiple phosphorylation sites on the AMPA receptor GluR1 subunit, Neuron, vol.16, pp.1179-88, 1996.

E. M. Rodrigues, S. L. Scudder, M. S. Goo, and G. N. Patrick, A?-Induced Synaptic Alterations Require the E3 Ubiquitin Ligase Nedd4-1, J Neurosci, vol.36, pp.1590-1595, 2016.

J. K. Ryu, T. Cho, H. B. Choi, Y. T. Wang, and J. G. Mclarnon, Microglial VEGF receptor response is an integral chemotactic component in Alzheimer's disease pathology, J Neurosci, vol.29, pp.3-13, 2009.

M. W. Salter and L. V. Kalia, Src kinases: a hub for NMDA receptor regulation, Nat Rev Neurosci, vol.5, pp.317-345, 2004.

S. B. Selleck, Signaling from across the way: transactivation of VEGF receptors by HSPGs, Mol Cell, vol.22, pp.431-433, 2006.

A. Serrano-pozo, M. P. Frosch, E. Masliah, and B. T. Hyman, Neuropathological alterations in Alzheimer disease, Cold Spring Harb Perspect Med, vol.1, p.6189, 2011.

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.2866-75, 2007.

G. M. Shankar, S. Li, T. H. Mehta, A. Garcia-munoz, N. E. Shepardson et al., Amyloid-beta protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory, Nat Med, vol.14, pp.837-879, 2008.

M. Sheng, B. L. Sabatini, and T. C. Südhof, Synapses and Alzheimer's disease, Cold Spring Harb Perspect Biol, vol.4, p.5777, 2012.

L. M. Smith and S. M. Strittmatter, Binding Sites for Amyloid-? Oligomers and Synaptic Toxicity, Cold Spring Harb Perspect Med, p.24075, 2017.

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.1051-1059, 2005.

W. B. Stine, K. N. Dahlgren, G. A. Krafft, and M. J. Ladu, In vitro characterization of conditions for amyloid-beta peptide oligomerization and fibrillogenesis, J Biol Chem, vol.278, pp.11612-11634, 2003.

T. Tada and M. Sheng, Molecular mechanisms of dendritic spine morphogenesis, Curr Opin Neurobiol, vol.16, pp.95-101, 2006.

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.1791-800, 2002.

T. Thomas, S. Miners, and S. Love, Post-mortem assessment of hypoperfusion of cerebral cortex in Alzheimer's disease and vascular dementia, Brain, vol.138, pp.1059-69, 2015.

T. Thomas, S. Miners, and S. Love, Post-mortem assessment of hypoperfusion of cerebral cortex in Alzheimer's disease and vascular dementia, Brain, vol.138, pp.1059-69, 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.

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

M. M. Verbeek, I. Otte-höller, J. Van-den-born, L. P. Van-den-heuvel, G. David et al., Agrin is a major heparan sulfate proteoglycan accumulating in Alzheimer's disease brain, Am J Pathol, vol.155, pp.2115-2140, 1999.

K. L. Viola and W. L. Klein, Amyloid ? oligomers in Alzheimer's disease pathogenesis, treatment, and diagnosis, Acta Neuropathol, vol.129, pp.183-206, 2015.

D. M. Walsh, I. Klyubin, J. V. Fadeeva, W. K. Cullen, R. Anwyl et al., Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo, Nature, vol.416, pp.535-544, 2002.

B. Winblad, P. Amouyel, S. Andrieu, C. Ballard, C. Brayne et al., Defeating Alzheimer's disease and other dementias: a priority for European science and society, Lancet Neurol, vol.5, pp.455-532, 2016.

S. P. Yang, D. G. Bae, H. J. Kang, B. J. Gwag, Y. S. Gho et al., Co-accumulation of vascular endothelial growth factor with beta-amyloid in the brain of patients with Alzheimer's disease, Neurobiol Aging, vol.25, pp.283-90, 2004.

S. P. Yang, B. O. Kwon, Y. S. Gho, and C. B. Chae, Specific interaction of VEGF165 with beta-amyloid, and its protective effect on beta-amyloid-induced neurotoxicity, J Neurochem, vol.93, pp.118-145, 2005.

W. Q. Zhao, F. Santini, R. Breese, D. Ross, X. D. Zhang et al., Inhibition of calcineurin-mediated endocytosis and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors prevents amyloid beta oligomer-induced synaptic disruption, J Biol Chem, vol.285, pp.7619-7651, 2010.

K. Aase, G. Euler, X. Li, A. Pontén, P. Thorén et al., Vascular Endothelial Growth Factor-B-Deficient Mice Display an Atrial Conduction Defect, Circulation, vol.104, pp.358-364, 2001.

G. Akiri, D. Nahari, Y. Finkelstein, S. Le, O. Elroy-stein et al., Regulation of vascular endothelial growth factor (VEGF) expression is mediated by internal initiation of translation and alternative initiation of transcription, Oncogene, vol.17, pp.227-236, 1998.

E. Alberdi, M. V. Sánchez-gómez, F. Cavaliere, A. Pérez-samartín, J. L. Zugaza et al., Amyloid beta oligomers induce Ca2+ dysregulation and neuronal death through activation of ionotropic glutamate receptors, Cell Calcium, vol.47, pp.264-272, 2010.

R. J. Albuquerque, T. Hayashi, W. G. Cho, M. E. Kleinman, S. Dridi et al., Alternatively spliced vascular endothelial growth factor receptor-2 is an essential endogenous inhibitor of lymphatic vessel growth, Nat. Med, vol.15, pp.1023-1030, 2009.

D. W. Ali and M. W. Salter, NMDA receptor regulation by Src kinase signalling in excitatory synaptic transmission and plasticity, vol.7, 2001.

C. G. Almeida, D. Tampellini, R. H. Takahashi, P. Greengard, M. T. Lin et al., Beta-amyloid accumulation in APP mutant neurons reduces PSD-95 and GluR1 in synapses, Neurobiol. Dis, vol.20, pp.187-198, 2005.

A. Altmann, L. Tian, V. W. Henderson, and M. D. Greicius, Sex modifies the APOErelated risk of developing Alzheimer disease, Annals of Neurology, vol.75, pp.563-573, 2014.

A. Álvarez-aznar, L. Muhl, and K. Gaengel, VEGF Receptor Tyrosine Kinases: Key Regulators of Vascular Function, Curr. Top. Dev. Biol, vol.123, pp.433-482, 2017.

, Alzheimer's disease facts and figures, Alzheimer's & Dementia, vol.14, pp.367-429, 2018.

N. Amada, K. Aihara, R. Ravid, and M. Horie, Reduction of NR1 and phosphorylated Ca2+/calmodulin-dependent protein kinase II levels in Alzheimer's disease, Neuroreport, vol.16, pp.1809-1813, 2005.

F. Amar, M. A. Sherman, T. Rush, M. Larson, G. Boyle et al., Amyloid-? oligomer A?*56 induces specific alterations of tau phosphorylation and neuronal signaling, Sci Signal, p.10, 2017.

A. Ambrosini, L. Bresciani, S. Fracchia, N. Brunello, and G. Racagni, Metabotropic glutamate receptors negatively coupled to adenylate cyclase inhibit N-methyl-Daspartate receptor activity and prevent neurotoxicity in mesencephalic neurons in vitro, Mol Pharmacol, vol.47, pp.1057-1064, 1995.

R. Anand, K. D. Gill, and A. A. Mahdi, Therapeutics of Alzheimer's disease: Past, present and future, Neuropharmacology, vol.76, pp.27-50, 2014.

A. Anoop, P. K. Singh, R. S. Jacob, and S. K. Maji, CSF Biomarkers for Alzheimer's Disease Diagnosis, Int J Alzheimers Dis, 2010.

D. Antequera, A. Portero, M. Bolos, G. Orive, R. M. Hernández et al., Encapsulated VEGF-Secreting Cells Enhance Proliferation of Neuronal Progenitors in the Hippocampus of A?PP/Ps1 Mice, Journal of Alzheimer's Disease, vol.29, pp.187-200, 2012.

J. I. Arellano, R. Benavides-piccione, J. Defelipe, and R. Yuste, Ultrastructure of Dendritic Spines: Correlation Between Synaptic and Spine Morphologies, Front Neurosci, vol.1, pp.131-143, 2007.

A. Asai, J. Qiu, Y. Narita, S. Chi, N. Saito et al., High Level Calcineurin Activity Predisposes Neuronal Cells to Apoptosis, J. Biol. Chem, vol.274, pp.34450-34458, 1999.

A. Attardo, J. E. Fitzgerald, and M. J. Schnitzer, Impermanence of dendritic spines in live adult CA1 hippocampus, Nature, vol.523, pp.592-596, 2015.

C. S. Atwood, R. N. Martins, M. A. Smith, and G. Perry, Senile plaque composition and posttranslational modification of amyloid-? peptide and associated proteins, Amyloid Peptides, vol.23, pp.1343-1350, 2002.

I. Avraham-davidi, Y. Ely, V. N. Pham, D. Castranova, M. Grunspan et al., ApoBcontaining lipoproteins regulate angiogenesis by modulating expression of VEGF receptor 1, Nat. Med, vol.18, pp.967-973, 2012.

L. W. Babus, E. M. Little, K. E. Keenoy, S. S. Minami, E. Chen et al., Decreased dendritic spine density and abnormal spine morphology in Fyn knockout mice, Brain Res, pp.96-102, 1415.

R. E. Bachelder, A. Crago, and J. Chung, Vascular endothelial growth factor is an autocrine survival factor for neuropilin-expressing breast carcinoma cells, Cancer Res, vol.61, pp.5736-5776, 2001.

R. E. Bachelder, M. A. Wendt, and A. M. Mercurio, Vascular endothelial growth factor promotes breast carcinoma invasion in an autocrine manner by regulating the chemokine receptor CXCR4, Cancer Res, vol.62, pp.7203-7209, 2002.

K. R. Bales, T. Verina, R. C. Dodel, Y. Du, L. Altstiel et al., Lack of apolipoprotein E dramatically reduces amyloid ?-peptide deposition, Nature Genetics, vol.17, p.263, 1997.

C. Bancher, C. Brunner, H. Lassmann, H. Budka, K. Jellinger et al., Accumulation of abnormally phosphorylated ? precedes the formation of neurofibrillary tangles in Alzheimer's disease, Brain Research, vol.477, pp.90-99, 1989.

K. Baranger, Y. Marchalant, A. E. Bonnet, N. Crouzin, A. Carrete et al., MT5-MMP is a new pro-amyloidogenic proteinase that promotes amyloid pathology and cognitive decline in a transgenic mouse model of Alzheimer's disease, Cell. Mol. Life Sci, vol.73, pp.217-236, 2016.

W. G. Barreto, G. C. Siufi, E. Y. Kimura, J. G. Pessoa, J. B. Pesqueiro et al., VEGF Expression as Prognostic Factor in Early Clinical Stages of Chronic Lymphocytic Leukemia Patients, Blood, vol.112, pp.4173-4173, 2008.

A. Barria, Regulatory Phosphorylation of AMPA-Type Glutamate Receptors by CaM-KII During Long-Term Potentiation, Science, vol.276, pp.2042-2045, 1997.

A. E. Barry, I. Klyubin, J. M. Mc-donald, A. J. Mably, M. A. Farrell et al., Alzheimer's Disease Brain-Derived Amyloid--Mediated Inhibition of LTP In Vivo Is Prevented by Immunotargeting Cellular Prion Protein, Journal of Neuroscience, vol.31, pp.7259-7263, 2011.

D. Basagiannis, S. Zografou, C. Murphy, T. Fotsis, L. Morbidelli et al., VEGF induces signalling and angiogenesis by directing VEGFR2 internalisation through macropinocytosis, Development, vol.143, 2016.

D. O. Bates, T. Cui, J. M. Doughty, M. Winkler, M. Sugiono et al., VEGF165b, an inhibitory splice variant of vascular endothelial growth factor, is down-regulated in renal cell carcinoma, Cancer Res, vol.62, pp.4123-4131, 2002.

A. Baude, Z. Nusser, E. Molnár, R. A. Mcilhinney, and P. Somogyi, High-resolution immunogold localization of AMPA type glutamate receptor subunits at synaptic and non-synaptic sites in rat hippocampus, Neuroscience, vol.69, pp.1031-1055, 1995.

V. L. Bautch, VEGF-Directed Blood Vessel Patterning: From Cells to Organism. Cold Spring Harb Perspect Med 2, 2012.

K. Bayer, P. De-koninck, A. S. Leonard, J. W. Hell, and H. Schulman, Interaction with the NMDA receptor locks CaMKII in an active conformation, Nature, vol.411, pp.801-805, 2001.

M. F. Bear and A. C. Wickliffe, Long-Term Depression in Hippocampus, Annu. Rev. Neurosc, vol.19, pp.437-62, 1996.

N. Beckouche, M. Bignon, V. Lelarge, T. Mathivet, C. Pichol-thievend et al., The interaction of heparan sulfate proteoglycans with endothelial transglutaminase-2 limits VEGF165-induced angiogenesis, Sci Signal, vol.8, p.70, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01184004

L. M. Bekris, C. Yu, T. D. Bird, and D. W. Tsuang, Genetics of Alzheimer Disease, J Geriatr Psychiatry Neurol, vol.23, pp.213-227, 2010.

R. Benavides-piccione, I. Fernaud-espinosa, V. Robles, R. Yuste, and J. Defelipe, Age-Based Comparison of Human Dendritic Spine Structure Using Complete ThreeDimensional Reconstructions, Cereb Cortex, vol.23, pp.1798-1810, 2013.

R. E. Bennett, S. L. Devos, S. Dujardin, B. Corjuc, R. Gor et al., Enhanced Tau Aggregation in the Presence of Amyloid ?, Am J Pathol, vol.187, pp.1601-1612, 2017.

A. Bernard and M. Khrestchatisky, Assessing the Extent of RNA Editing in the TMII Regions of GluR5 and GluR6 Kainate Receptors During Rat Brain Development, Journal of Neurochemistry, vol.62, pp.2057-2060, 1994.

K. P. Berry and E. Nedivi, Spine dynamics: Are they all the same?, Neuron, vol.96, pp.43-55, 2017.

K. Bhaskar, G. A. Hobbs, S. Yen, and G. Lee, Tyrosine phosphorylation of tau accompanies disease progression in transgenic mouse models of tauopathy, Neuropathol. Appl. Neurobiol, vol.36, pp.462-477, 2010.

M. Bhattacharyya, A. Nandanoor, M. Osman, C. Kasinathan, and P. Frederikse, Erratum to: NMDA Glutamate Receptor NR1, NR2A and NR2B Expression and NR2B Tyr-1472 Phosphorylation in the Lens, Neurochemical Research, vol.39, pp.1833-1833, 2014.

C. Bidoret, A. Ayon, B. Barbour, and M. Casado, Presynaptic NR2A-containing NMDA receptors implement a high-pass filter synaptic plasticity rule, vol.106, pp.14126-14131, 2009.

M. L. Billingsley, C. Ellis, R. L. Kincaid, J. Martin, M. L. Schmidt et al., Calcineurin Immunoreactivity in Alzheimer's Disease, Experimental Neurology, vol.126, pp.178-184, 1994.

R. Binétruy-tournaire, C. Demangel, B. Malavaud, R. Vassy, S. Rouyre et al., Identification of a peptide blocking vascular endothelial growth factor (VEGF)-mediated angiogenesis, EMBO J, vol.19, pp.1525-1533, 2000.

T. D. Bird, Genetic Aspects of Alzheimer Disease, Genet Med, vol.10, pp.231-239, 2008.

T. V. Bliss and G. L. Collingridge, A synaptic model of memory: long-term potentiation in the hippocampus, Nature, vol.361, pp.31-39, 1993.

B. L. Bloodgood, A. J. Giessel, and B. L. Sabatini, Biphasic Synaptic Ca Influx Arising from Compartmentalized Electrical Signals in Dendritic Spines, PLOS Biology, vol.7, p.1000190, 2009.

T. Bonhoeffer and R. Yuste, Spine Motility: Phenomenology, Mechanisms, and Function. Neuron, vol.35, pp.1019-1027, 2002.

Z. A. Bortolotto, Z. I. Bashir, C. H. Davies, and G. L. Collingridge, A molecular switch activated by metabotropic glutamate receptors regulates induction of long-term potentiation, Nature, vol.368, pp.740-743, 1994.

J. Bourne and K. M. Harris, Do thin spines learn to be mushroom spines that remember?, Curr. Opin. Neurobiol, vol.17, pp.381-386, 2007.

G. Bouvier, C. Bidoret, M. Casado, and P. Paoletti, Presynaptic NMDA receptors: Roles and rules, Neuroscience, vol.311, pp.322-340, 2015.

H. Braak and E. Braak, Neuropathological stageing of Alzheimer-related changes, Acta Neuropathol, vol.82, issue.4, pp.239-59, 1991.

S. P. Braithwaite, J. B. Stock, P. J. Lombroso, and A. C. Nairn, Protein Phosphatases and Alzheimer's Disease. Prog, Mol. Biol. Transl. Sci, vol.106, pp.343-379, 2012.

G. Breier, U. Albrecht, S. Sterrer, and W. Risau, Expression of vascular endothelial growth factor during embryonic angiogenesis and endothelial cell differentiation, Development, vol.114, issue.2, pp.521-553, 1992.

V. Bruno, A. Copani, T. Knöpfel, R. Kuhn, G. Casabona et al., Activation of metabotropic glutamate receptors coupled to inositol phospholipid hydrolysis amplifies NMDA-induced neuronal degeneration in cultured cortical cells, Neuropharmacology, vol.34, pp.1089-1098, 1995.

A. Buisson, S. P. Yu, and D. W. Choi, DCG-IV Selectively Attenuates Rapidly Triggered NMDA-induced Neurotoxicity in Cortical Neurons, European Journal of Neuroscience, vol.8, pp.138-143, 1996.

R. Cacace, K. Sleegers, and C. Van-broeckhoven, Molecular genetics of early-onset Alzheimer's disease revisited, Alzheimers Dement, vol.12, pp.733-748, 2016.

A. Caccamo, S. Oddo, L. M. Billings, K. N. Green, H. Martinez-coria et al., M1 Receptors Play a Central Role in Modulating AD-like Pathology in Transgenic Mice, Neuron, vol.49, pp.671-682, 2006.

M. Cammalleri, D. Martini, C. Ristori, A. M. Timperio, and P. Bagnoli, Vascular endothelial growth factor up-regulation in the mouse hippocampus and its role in the control of epileptiform activity, European Journal of Neuroscience, vol.33, pp.482-498, 2011.

D. Campion, C. Dumanchin, D. Hannequin, B. Dubois, S. Belliard et al., , 1999.

, Early-onset autosomal dominant Alzheimer disease: prevalence, genetic heterogeneity, and mutation spectrum, Am J Hum Genet, vol.65, pp.664-670

L. Cao, X. Jiao, D. S. Zuzga, Y. Liu, D. M. Fong et al., VEGF links hippocampal activity with neurogenesis, learning and memory, Nature Genetics, vol.36, pp.827-835, 2004.

X. Cao and T. C. Südhof, A transcriptionally [correction of transcriptively] active complex of APP with Fe65 and histone acetyltransferase Tip60, Science, vol.293, pp.115-120, 2001.

A. Cariboni, K. Davidson, E. Dozio, F. Memi, Q. Schwarz et al., VEGF signalling controls GnRH neuron survival via NRP1 independently of KDR and blood vessels, Development, p.3723, 2011.

P. Carmeliet, P. Carmeliet, V. Ferreira, G. Breier, S. Pollefeyt et al., Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele, Nature Med, vol.9, pp.435-439, 1996.

P. Carmeliet and R. K. Jain, Molecular mechanisms and clinical applications of angiogenesis, Nature, vol.473, pp.298-307, 2011.

P. Carmeliet and C. Ruiz-de-almodovar, VEGF ligands and receptors: implications in neurodevelopment and neurodegeneration, Cellular and Molecular Life Sciences, vol.70, pp.1763-1778, 2013.

P. Carmeliet and M. Tessier-lavigne, Common mechanisms of nerve and blood vessel wiring, Nature, vol.436, pp.193-200, 2005.

R. C. Carroll, E. C. Beattie, M. Von-zastrow, and R. C. Malenka, Role of AMPA receptor endocytosis in synaptic plasticity, Nat. Rev. Neurosci, vol.2, pp.315-324, 2001.

R. V. Carvalho, F. Da-silva-ferreira, L. Heimfarth, P. Pierozan, C. Fernandes et al., Acute Hyperammonemia Induces NMDA-Mediated Hypophosphorylation of Intermediate Filaments Through PP1 and PP2B in Cerebral Cortex of Young Rats, Neurotoxicity Research, vol.30, pp.138-149, 2016.

C. V. Cauwenberghe, C. V. Broeckhoven, and K. Sleegers, The genetic landscape of Alzheimer disease: clinical implications and perspectives, Genetics in Medicine, vol.18, pp.421-430, 2016.

V. Cavallucci, N. Berretta, A. Nobili, R. Nisticò, N. B. Mercuri et al., Calcineurin Inhibition Rescues Early Synaptic Plasticity Deficits in a Mouse Model of Alzheimer's Disease, Neuromol Med, vol.15, pp.541-548, 2013.

C. Suarez, S. Pieren, M. Cariolato, L. Arn, S. Hoffmann et al., A VEGF-A splice variant defective for heparan sulfate and neuropilin-1 binding shows attenuated signaling through VEGFR-2, Cellular and Molecular Life Sciences, vol.63, pp.2067-2077, 2006.

A. Chakraborty, M. Chatterjee, H. Twaalfhoven, M. Del-campo-milan, C. E. Teunissen et al., Vascular Endothelial Growth Factor remains unchanged in cerebrospinal fluid of patients with Alzheimer's disease and vascular dementia, vol.10, p.58, 2018.

C. Chang, J. R. Neilson, J. H. Bayle, J. E. Gestwicki, A. Kuo et al., A Field of Myocardial-Endocardial NFAT Signaling Underlies Heart Valve Morphogenesis, vol.118, pp.649-663, 2004.

S. Chasseigneaux and B. Allinquant, Functions of A?, sAPP? and sAPP? : similarities and differences, J. Neurochem, vol.120, issue.1, pp.99-108, 2012.

S. Chauvet, K. Burk, and F. Mann, Navigation rules for vessels and neurons: cooperative signaling between VEGF and neural guidance cues, Cell. Mol. Life Sci, vol.70, pp.1685-1703, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00862136

B. Chen and K. W. Roche, Regulation of NMDA Receptors by Phosphorylation, Neuropharmacology, vol.53, pp.362-368, 2007.

J. Chen, P. R. Somanath, O. Razorenova, W. S. Chen, N. Hay et al., Akt1 regulates pathological angiogenesis, vascular maturation and permeability in vivo, Nat Med, vol.11, pp.1188-1196, 2005.

L. Chen, Y. Wang, J. Chen, and G. Tseng, NMDA receptor triggered molecular cascade underlies compression-induced rapid dendritic spine plasticity in cortical neurons, Exp. Neurol, vol.266, pp.86-98, 2015.

Q. Chen, W. Wei, T. Shimahara, and C. Xie, Alzheimer Amyloid ?-Peptide Inhibits the Late Phase of Long-Term Potentiation through CalcineurinDependent Mechanisms in the Hippocampal Dentate Gyrus, Neurobiology of Learning and Memory, vol.77, pp.354-371, 2002.
URL : https://hal.archives-ouvertes.fr/hal-00201234

X. Chen, J. M. Levy, A. Hou, C. Winters, R. Azzam et al., PSD-95 family MAGUKs are essential for anchoring AMPA and NMDA receptor complexes at the postsynaptic density, Proc. Natl. Acad. Sci, vol.112, pp.6983-6992, 2015.

G. Chêne, A. Beiser, R. Au, S. R. Preis, P. A. Wolf et al., Gender and incidence of dementia in the Framingham Heart Study from mid-adult life, Alzheimer's & Dementia, vol.11, pp.310-320, 2015.

I. H. Cheng, K. Scearce-levie, J. Legleiter, J. J. Palop, H. Gerstein et al., Accelerating Amyloid-? Fibrillization Reduces Oligomer Levels and Functional Deficits in Alzheimer Disease Mouse Models, J. Biol. Chem, vol.282, pp.23818-23828, 2007.

M. Chiappelli, B. Borroni, S. Archetti, E. Calabrese, M. M. Corsi et al., VEGF gene and phenotype relation with Alzheimer's disease and mild cognitive impairment, Rejuvenation Res, vol.9, pp.485-493, 2006.

P. Chiodelli, S. Mitola, C. Ravelli, P. Oreste, M. Rusnati et al., Heparan sulfate proteoglycans mediate the angiogenic activity of the vascular endothelial growth factor receptor-2 agonist gremlin, Arterioscler. Thromb. Vasc. Biol, vol.31, pp.116-127, 2011.

S. Cho, M. H. Park, C. Han, K. Yoon, and Y. H. Koh, VEGFR2 alteration in Alzheimer's disease, vol.7, p.17713, 2017.

C. H. Choi, S. Y. Song, J. Choi, Y. Park, H. Kang et al., Prognostic significance of VEGF expression in patients with bulky cervical carcinoma undergoing neoadjuvant chemotherapy, BMC Cancer, vol.8, p.295, 2008.

B. A. Chromy, R. J. Nowak, M. P. Lambert, K. L. Viola, L. Chang et al., Self-assembly of Abeta(1-42) into globular neurotoxins, Biochemistry, vol.42, pp.12749-12760, 2003.

Z. Chu and J. J. Hablitz, Quisqualate induces an inward current via mGluR activation in neocortical pyramidal neurons, Brain Research, vol.879, issue.1-2, pp.88-92, 2000.

M. Cissé, B. Halabisky, J. Harris, N. Devidze, D. B. Dubal et al., Reversing EphB2 depletion rescues cognitive functions in Alzheimer model, Nature, vol.469, pp.47-52, 2011.

M. Citron, T. Oltersdorf, C. Haass, L. Mcconlogue, A. Y. Hung et al., Mutation of the beta-amyloid precursor protein in familial Alzheimer's disease increases beta-protein production, Nature, vol.360, pp.672-674, 1992.

R. F. Clark, M. Hutton, M. Fuldner, S. Froelich, E. Karran et al., The structure of the presenilin 1 (S182) gene and identification of six novel mutations in early onset AD families, vol.11, pp.219-222, 1995.

J. P. Cleary, D. M. Walsh, J. J. Hofmeister, G. M. Shankar, M. A. Kuskowski et al., Natural oligomers of the amyloid-beta protein specifically disrupt cognitive function, Nat. Neurosci, vol.8, pp.79-84, 2005.

N. A. Clipstone and G. R. Crabtree, Identification of calcineurin as a key signalling enzyme in T-lymphocyte activation, Nature, vol.357, pp.695-697, 1992.

C. Coisne and B. Engelhardt, Tight junctions in brain barriers during central nervous system inflammation, Antioxid. Redox Signal, vol.15, pp.1285-1303, 2011.

P. A. Colley, F. S. Sheu, and A. Routtenberg, Inhibition of protein kinase C blocks two components of LTP persistence, leaving initial potentiation intact, J. Neurosci, vol.10, pp.3353-3360, 1990.

D. T. Connolly, D. M. Heuvelman, R. Nelson, J. V. Olander, B. L. Eppley et al., Tumor vascular permeability factor stimulates endothelial cell growth and angiogenesis, J. Clin. Invest, vol.84, pp.1470-1478, 1989.

A. Contractor, C. Mulle, and G. T. Swanson, Kainate receptors coming of age: milestones of two decades of research, Trends in Neurosciences, vol.34, pp.154-163, 2011.

S. J. Coultrap, R. K. Freund, H. O'leary, J. L. Sanderson, K. W. Roche et al., Autonomous CaMKII mediates both LTP and LTD using a mechanism for differential substrate site selection, Cell Rep, vol.6, pp.431-437, 2014.

M. J. Cross, J. Dixelius, T. Matsumoto, and L. Claesson-welsh, VEGF-receptor signal transduction, Trends in Biochemical Sciences, vol.28, pp.488-494, 2003.

M. Cruts and C. Van-broeckhoven, Presenilin mutations in Alzheimer's disease, Hum. Mutat, vol.11, pp.183-190, 1998.

J. A. Cummings, R. M. Mulkey, R. A. Nicoll, and R. C. Malenka, Ca2+ Signaling Requirements for Long-Term Depression in the Hippocampus, Neuron, vol.16, pp.825-833, 1996.

M. D'amelio, V. Cavallucci, S. Middei, C. Marchetti, S. Pacioni et al., Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer's disease, Nat Neurosci, vol.14, pp.69-76, 2011.

M. D'amelio, M. Sheng, and F. Cecconi, Caspase-3 in the central nervous system: beyond apoptosis, Trends in Neurosciences, vol.35, pp.700-709, 2012.

K. N. Dahlgren, A. M. Manelli, W. B. Stine, L. K. Baker, G. A. Krafft et al., Oligomeric and Fibrillar Species of Amyloid-? Peptides Differentially Affect Neuronal Viability, J. Biol. Chem, vol.277, pp.32046-32053, 2002.

P. De-rossi, E. Harde, J. P. Dupuis, L. Martin, N. Chounlamountri et al., A critical role for VEGF and VEGFR2 in NMDA receptor synaptic function and fear-related behavior, Molecular Psychiatry, vol.21, pp.1768-1780, 2016.

A. De-simoni, C. B. Griesinger, and F. A. Edwards, Development of rat CA1 neurones in acute versus organotypic slices: role of experience in synaptic morphology and activity, J. Physiol, vol.550, pp.135-147, 2003.

Y. Deng, D. Atri, A. Eichmann, and M. Simons, Endothelial ERK signaling controls lymphatic fate specification, J Clin Invest, vol.123, pp.1202-1215, 2013.
DOI : 10.1172/jci63034

URL : http://europepmc.org/articles/pmc3582116?pdf=render

Y. Deng, X. Zhang, and M. Simons, Molecular controls of lymphatic VEGFR3 signaling, Arterioscler. Thromb. Vasc. Biol, vol.35, pp.421-429, 2015.

V. Derkach, A. Barria, and T. R. Soderling, Ca2+/calmodulin-kinase II enhances channel conductance of ?-amino-3-hydroxy-5-methyl-4-isoxazolepropionate type glutamate receptors, vol.96, pp.3269-3274, 1999.

G. H. Diering, A. S. Gustina, and R. L. Huganir, PKA-GluA1 coupling via AKAP5 controls AMPA receptor phosphorylation and cell-surface targeting during bidirectional homeostatic plasticity, Neuron, vol.84, pp.790-805, 2014.

K. T. Dineley, D. Hogan, W. Zhang, and G. Taglialatela, Acute Inhibition Of Calcineurin Restores Associative Learning And Memory In Tg2576 APP Transgenic Mice, Neurobiol. Learn. Mem, vol.88, pp.217-224, 2007.

K. T. Dineley, R. Kayed, V. Neugebauer, Y. Fu, W. Zhang et al., Amyloid-? oligomers impair fear conditioned memory in a calcineurindependent fashion in mice, Journal of Neuroscience Research, vol.88, issue.13, pp.2923-2932, 2010.

X. Ding, R. Gu, M. Zhang, H. Ren, Q. Shu et al., Microglia enhanced the angiogenesis, migration and proliferation of co-cultured RMECs, BMC Ophthalmology, p.18, 2018.

J. Disalvo, M. L. Bayne, G. Conn, P. W. Kwok, P. G. Trivedi et al., Purification and Characterization of a Naturally Occurring Vascular Endothelial Growth Factor · Placenta Growth Factor Heterodimer, J. Biol. Chem, vol.270, pp.7717-7723, 1995.

R. S. Doody, R. G. Thomas, M. Farlow, T. Iwatsubo, B. Vellas et al., Phase 3 Trials of Solanezumab for Mild-to-Moderate Alzheimer's Disease, New England Journal of Medicine, vol.370, pp.311-321, 2014.

M. M. Dorostkar, C. Zou, L. Blazquez-llorca, and J. Herms, Analyzing dendritic spine pathology in Alzheimer's disease: problems and opportunities, Acta Neuropathol, vol.130, pp.1-19, 2015.

M. Dougher and B. I. Terman, Autophosphorylation of KDR in the kinase domain is required for maximal VEGF-stimulated kinase activity and receptor internalization, Oncogene, vol.18, p.1619, 1999.

G. Drewes, E. Mandelkow, K. Baumann, J. Goris, W. Merlevede et al., Dephosphorylation of tau protein and Alzheimer paired helical filaments by calcmeurin and phosphatase2A, FEBS Letters, vol.336, pp.425-432, 1994.

F. Dulin, F. Léveillé, J. B. Ortega, J. Mornon, A. Buisson et al., p3 peptide, a truncated form of A? devoid of synaptotoxic effect, does not assemble into soluble oligomers, FEBS Letters, vol.582, pp.1865-1870, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00279701

D. J. Dumont, L. Jussila, J. Taipale, A. Lymboussaki, T. Mustonen et al., Cardiovascular failure in mouse embryos deficient in VEGF receptor-3, Science, vol.282, pp.946-949, 1998.

J. P. Dupuis, L. Ladépêche, H. Seth, L. Bard, J. Varela et al., Surface dynamics of GluN2B-NMDA receptors controls plasticity of maturing glutamate synapses, EMBO J, vol.33, pp.842-861, 2014.

S. Ebrahimi and S. Okabe, Structural dynamics of dendritic spines: Molecular composition, geometry and functional regulation, Biochimica et Biophysica Acta (BBA) -Biomembranes, pp.2391-2398, 1838.
DOI : 10.1016/j.bbamem.2014.06.002

URL : https://doi.org/10.1016/j.bbamem.2014.06.002

N. J. Emptage, C. A. Reid, and A. Fine, Calcium Stores in Hippocampal Synaptic Boutons Mediate Short-Term Plasticity, Store-Operated Ca2+ Entry, and Spontaneous Transmitter Release, Neuron, vol.29, pp.197-208, 2001.

T. Endoh, Characterization of modulatory effects of postsynaptic metabotropic glutamate receptors on calcium currents in rat nucleus tractus solitarius, Brain Research, vol.1024, pp.212-224, 2004.

F. Engert and T. Bonhoeffer, Dendritic spine changes associated with hippocampal longterm synaptic plasticity, Nature, vol.399, pp.66-70, 1999.

L. Erskine, S. Reijntjes, T. Pratt, L. Denti, Q. Schwarz et al., VEGF signaling through neuropilin 1 guides commissural axon crossing at the optic chiasm, Neuron, vol.70, pp.951-965, 2011.
DOI : 10.1016/j.neuron.2011.02.052

URL : https://doi.org/10.1016/j.neuron.2011.02.052

G. Fearnley, G. Smith, M. Harrison, S. Wheatcroft, D. Tomlinson et al., Vascular endothelial growth factor-A regulation of blood vessel sprouting in health and disease, OA Biochemistry, vol.1, issue.1, p.5, 2013.

G. W. Fearnley, G. A. Smith, I. Abdul-zani, N. Yuldasheva, N. A. Mughal et al., VEGF-A isoforms program differential VEGFR2 signal transduction, trafficking and proteolysis, Biol Open, vol.5, pp.571-583, 2016.

I. M. Ferrando, R. Chaerkady, J. Zhong, H. Molina, H. K. Jacob et al., Identification of Targets of c-Src Tyrosine Kinase by Chemical Complementation and Phosphoproteomics, Mol Cell Proteomics, vol.11, pp.355-369, 2012.

N. Ferrara, K. Carver-moore, H. Chen, M. Dowd, L. Lu et al., Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene, Nature, vol.380, pp.439-442, 1996.

J. S. Ferreira, T. Papouin, L. Ladépêche, A. Yao, V. C. Langlais et al., , 2017.

, Co-agonists differentially tune GluN2B-NMDA receptor trafficking at hippocampal synapses, vol.6, p.25492, 2009.

J. C. Fiala, M. Feinberg, V. Popov, and K. M. Harris, Synaptogenesis via dendritic filopodia in developing hippocampal area CA1, J Neurosci, 1998.

M. Fischer, S. Kaech, U. Wagner, H. Brinkhaus, and A. Matus, Glutamate receptors regulate actin-based plasticity in dendritic spines, Nature Neuroscience, vol.3, pp.887-894, 2000.

M. F. Folstein, S. E. Folstein, and P. R. Mchugh, Mini-mental state": A practical method for grading the cognitive state of patients for the clinician, Journal of Psychiatric Research, vol.12, pp.189-198, 1975.

G. H. Fong, J. Rossant, M. Gertsenstein, and M. L. Breitman, Role of the Flt-1 receptor tyrosine kinase in regulating the assembly of vascular endothelium, Nature, vol.376, pp.66-70, 1995.

P. Formichi, C. Battisti, E. Radi, and A. Federico, Cerebrospinal fluid tau, A beta, and phosphorylated tau protein for the diagnosis of Alzheimer's disease, J. Cell. Physiol, vol.208, pp.39-46, 2006.

F. Forstreuter, R. Lucius, and R. Mentlein, Vascular endothelial growth factor induces chemotaxis and proliferation of microglial cells, Journal of Neuroimmunology, vol.132, pp.93-98, 2002.

T. C. Foster, K. M. Sharrow, J. R. Masse, C. M. Norris, and A. Kumar, Calcineurin Links Ca2+ Dysregulation with Brain Aging, J Neurosci, vol.21, pp.4066-4073, 2001.

M. L. Frandemiche, S. D. 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, vol.34, pp.6084-6097, 2014.

D. B. Freir, A. J. Nicoll, I. Klyubin, S. Panico, J. M. Mc-donald et al., Interaction between prion protein and toxic amyloid ? assemblies can be therapeutically targeted at multiple sites, Nature Communications, vol.2, p.336, 2011.

R. Frischknecht, M. Heine, D. Perrais, C. I. Seidenbecher, D. Choquet et al., Brain extracellular matrix affects AMPA receptor lateral mobility and shortterm synaptic plasticity, Nat. Neurosci, vol.12, pp.897-904, 2009.

H. Funato, M. Yoshimura, K. Kusui, A. Tamaoka, K. Ishikawa et al., Quantitation of amyloid beta-protein (A beta) in the cortex during aging and in Alzheimer's disease, Am J Pathol, vol.152, pp.1633-1640, 1998.

A. Gampel, L. Moss, M. C. Jones, V. Brunton, J. C. Norman et al., VEGF regulates the mobilization of VEGFR2/KDR from an intracellular endothelial storage compartment, Blood, vol.108, pp.2624-2631, 2006.

H. Gerber, A. Mcmurtrey, J. Kowalski, M. Yan, B. A. Keyt et al., Vascular Endothelial Growth Factor Regulates Endothelial Cell Survival through the Phosphatidylinositol 3?-Kinase/Akt Signal Transduction Pathway Requirement For Flk-1/Kdr Activation, J. Biol. Chem, vol.273, pp.30336-30343, 1998.

H. Gerhardt, VEGF and endothelial guidance in angiogenic sprouting, Organogenesis, vol.4, pp.241-246, 2008.

P. Giannakopoulos, F. R. Herrmann, T. Bussière, C. Bouras, E. Kövari et al., Tangle and neuron numbers, but not amyloid load, predict cognitive status in Alzheimer's disease, Neurology, vol.60, pp.1495-1500, 2003.

K. P. Giese, N. B. Fedorov, R. K. Filipkowski, and A. J. Silva, Autophosphorylation at Thr286 of the alpha calcium-calmodulin kinase II in LTP and learning, Science, vol.279, pp.870-873, 1998.

D. A. Gimbel, H. B. Nygaard, E. E. Coffey, E. C. Gunther, J. Laurén et al., Memory impairment in transgenic Alzheimer mice requires cellular prion protein, J. Neurosci, vol.30, pp.6367-6374, 2010.

C. Glabe, Structural Classification of Toxic Amyloid Oligomers, J Biol Chem, vol.283, pp.29639-29643, 2008.

Z. Gluzman-poltorak, T. Cohen, Y. Herzog, and G. Neufeld, Neuropilin-2 and Neuropilin-1 Are Receptors for the 165-Amino Acid Form of Vascular Endothelial Growth Factor (VEGF) and of Placenta Growth Factor-2, but Only Neuropilin-2 Functions as a Receptor for the 145-Amino Acid Form of VEGF, J. Biol. Chem, vol.275, pp.18040-18045, 2000.

C. S. Goodman and C. J. Shatz, Developmental mechanisms that generate precise patterns of neuronal connectivity, Cell, vol.72, pp.77-98, 1993.
DOI : 10.1016/s0092-8674(05)80030-3

S. G. Grant, T. J. O'dell, K. A. Karl, P. L. Stein, P. Soriano et al., Impaired long-term potentiation, spatial learning, and hippocampal development in fyn mutant mice, Science, vol.258, pp.1903-1910, 1992.
DOI : 10.1126/science.1361685

M. Graupera, J. Guillermet-guibert, L. C. Foukas, L. Phng, R. J. Cain et al., Angiogenesis selectively requires the p110alpha isoform of PI3K to control endothelial cell migration, Nature, vol.453, pp.662-666, 2008.

E. G. Gray, Axo-somatic and axo-dendritic synapses of the cerebral cortex, J Anat, vol.93, pp.420-433, 1959.

L. Groc, B. Gustafsson, and E. Hanse, AMPA signalling in nascent glutamatergic synapses: there and not there, Trends in Neurosciences, vol.29, pp.132-139, 2006.

L. Groc, M. Heine, L. Cognet, K. Brickley, F. A. Stephenson et al., Differential activity-dependent regulation of the lateral mobilities of AMPA and NMDA receptors, Nature Neuroscience, vol.7, pp.695-696, 2004.
URL : https://hal.archives-ouvertes.fr/hal-01551916

L. Groc, M. Heine, S. L. Cousins, F. A. Stephenson, B. Lounis et al., NMDA receptor surface mobility depends on NR2A-2B subunits, vol.103, pp.18769-18774, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01552913

A. Grothey and E. Galanis, Targeting angiogenesis: progress with anti-VEGF treatment with large molecules, Nature Reviews Clinical Oncology, vol.6, pp.507-518, 2009.

I. Grundke-iqbal, K. Iqbal, L. George, Y. C. Tung, K. S. Kim et al., Amyloid protein and neurofibrillary tangles coexist in the same neuron in Alzheimer disease, Proc. Natl. Acad. Sci. U.S.A, vol.86, pp.2853-2857, 1989.

F. S. Grünewald, A. E. Prota, A. Giese, and K. Ballmer-hofer, Structure-function analysis of VEGF receptor activation and the role of coreceptors in angiogenic signaling, Biochim. Biophys. Acta, vol.1804, pp.567-580, 2010.

Z. Gu, W. Liu, and Z. Yan, ?-Amyloid Impairs AMPA Receptor Trafficking and Function by Reducing Ca 2+ /Calmodulin-dependent Protein Kinase II Synaptic Distribution, Journal of Biological Chemistry, vol.284, pp.10639-10649, 2009.

M. L. Guzmán-hernández, G. Potter, K. Egervári, J. Z. Kiss, and T. Balla, Secretion of VEGF-165 has unique characteristics, including shedding from the plasma membrane, Mol Biol Cell, vol.25, pp.1061-1072, 2014.

C. Haass, C. Kaether, G. Thinakaran, and S. Sisodia, Trafficking and Proteolytic Processing of APP. Cold Spring Harb Perspect Med, vol.2, 2012.

C. E. Hagberg, A. Falkevall, X. Wang, E. Larsson, J. Huusko et al., Vascular endothelial growth factor B controls endothelial fatty acid uptake, Nature, vol.464, pp.917-921, 2010.
DOI : 10.1038/nature08945

J. J. Haigh, P. I. Morelli, H. Gerhardt, K. Haigh, J. Tsien et al., , 2003.

, Cortical and retinal defects caused by dosage-dependent reductions in VEGF-A paracrine signaling, Developmental Biology, vol.262, pp.225-241

B. J. Hall and A. Ghosh, Regulation of AMPA receptor recruitment at developing synapses, Trends in Neurosciences, vol.31, pp.82-89, 2008.

S. J. Harper and D. O. Bates, VEGF-A splicing: the key to anti-angiogenic therapeutics?, Nature reviews. Cancer, vol.8, p.880, 2008.

K. M. Harris, F. E. Jensen, and B. Tsao, Three-dimensional structure of dendritic spines and synapses in rat hippocampus (CA1) at postnatal day 15 and adult ages: implications for the maturation of synaptic physiology and long-term potentiation, J. Neurosci, vol.12, pp.2685-2705, 1992.

E. M. Hartenbach, T. A. Olson, J. J. Goswitz, D. Mohanraj, L. B. Twiggs et al., Vascular endothelial growth factor (VEGF) expression and survival in human epithelial ovarian carcinomas, Cancer Lett, vol.121, pp.169-175, 1997.

R. J. Harvey, M. Skelton-robinson, and M. N. Rossor, The prevalence and causes of dementia in people under the age of 65 years, Neurosurgery & Psychiatry, vol.74, pp.1206-1209, 2003.

Y. Hata, Molecular mechanism of the assembly of neurotransmitter receptors and cell adhesion molecules at postsynaptic density, 1998.

, Seikagaku, vol.70, pp.1414-1418

L. J. Hawinkels, K. Zuidwijk, H. W. Verspaget, E. S. De-jonge-muller, W. Van-duijn et al., VEGF release by MMP-9 mediated heparan sulphate cleavage induces colorectal cancer angiogenesis, Eur. J. Cancer, vol.44, pp.1904-1913, 2008.

Y. Hayashi, S. Shi, J. A. Esteban, A. Piccini, J. Poncer et al., Driving AMPA Receptors into Synapses by LTP and CaMKII: Requirement for GluR1 and PDZ Domain Interaction, Science, vol.287, pp.2262-2267, 2000.

Y. He, M. Zheng, Y. Ma, X. Han, X. Ma et al., Soluble oligomers and fibrillar species of amyloid ?-peptide differentially affect cognitive functions and hippocampal inflammatory response, Biochemical and Biophysical Research Communications, vol.429, pp.125-130, 2012.

Z. He, J. L. Guo, J. D. Mcbride, S. Narasimhan, H. Kim et al., Amyloid-? plaques enhance Alzheimer's brain tau-seeded pathologies by facilitating neuritic plaque tau aggregation, Nat. Med, vol.24, pp.29-38, 2018.

M. T. Heneka, M. J. Carson, J. E. Khoury, G. E. Landreth, F. Brosseron et al., Neuroinflammation in Alzheimer's disease, vol.14, pp.388-405, 2015.

E. Hinoi, K. Ogita, Y. Takeuchi, H. Ohashi, T. Maruyama et al., Characterization with [3H]quisqualate of group I metabotropic glutamate receptor subtype in rat central and peripheral excitable tissues, Neurochemistry International, vol.38, issue.3, pp.277-85, 2001.

S. Hiratsuka, O. Minowa, J. Kuno, T. Noda, and M. Shibuya, Flt-1 lacking the tyrosine kinase domain is sufficient for normal development and angiogenesis in mice, p.9349, 1998.

S. Hiratsuka, K. Nakao, K. Nakamura, M. Katsuki, Y. Maru et al., Membrane fixation of vascular endothelial growth factor receptor 1 ligand-binding domain is important for vasculogenesis and angiogenesis in mice, Mol. Cell. Biol, vol.25, pp.346-354, 2005.

T. J. Hohman, S. P. Bell, and A. L. Jefferson, The role of vascular endothelial growth factor in neurodegeneration and cognitive decline: exploring interactions with biomarkers of Alzheimer disease, JAMA Neurol, vol.72, pp.520-529, 2015.

M. Hollmann, M. Hartley, and S. Heinemann, Ca2+ permeability of KA-AMPA--gated glutamate receptor channels depends on subunit composition, Science, vol.252, pp.851-853, 1991.

M. Hollmann and S. Heinemann, Cloned Glutamate Receptors, Annual Review of Neuroscience, vol.17, pp.31-108, 1994.

D. I. Holmes, Z. , and I. , The vascular endothelial growth factor (VEGF) family: angiogenic factors in health and disease, Genome Biol, vol.6, p.209, 2005.

K. Holmes, E. Chapman, V. See, and M. J. Cross, VEGF Stimulates RCAN1.4 Expression in Endothelial Cells via a Pathway Requiring Ca2+/Calcineurin and Protein Kinase C-?, PLoS One, vol.5, p.11435, 2010.

K. Holmes, O. L. Roberts, A. M. Thomas, and M. J. Cross, Vascular endothelial growth factor receptor-2: Structure, function, intracellular signalling and therapeutic inhibition, Cellular Signalling, vol.19, 2003.

C. C. Hong, Q. P. Peterson, J. Hong, and R. T. Peterson, Artery/Vein Specification Is Governed by Opposing Phosphatidylinositol-3 Kinase and MAP Kinase/ERK Signaling, Curr Biol, vol.16, pp.1366-1372, 2006.

S. Hong, V. F. Beja-glasser, B. M. Nfonoyim, A. Frouin, S. Li et al., Complement and microglia mediate early synapse loss in Alzheimer mouse models, Science, p.8373, 2016.

N. Honkura, M. Matsuzaki, J. Noguchi, G. C. Ellis-davies, and H. Kasai, The subspine organization of actin fibers regulates the structure and plasticity of dendritic spines, Neuron, vol.57, pp.719-729, 2008.

C. H. Horner and E. Arbuthnott, Methods of estimation of spine density--are spines evenly distributed throughout the dendritic field?, J Anat, vol.177, pp.179-184, 1991.

C. Hornig and H. A. Weich, Soluble VEGF receptors, Angiogenesis, vol.3, pp.33-39, 1999.

K. Hsiao, P. Chapman, S. Nilsen, C. Eckman, Y. Harigaya et al., Correlative Memory Deficits, AP3 Elevation, and Amyloid Plaques in Transgenic Mice, vol.274, p.5, 1996.

H. Hsieh, J. Boehm, C. Sato, T. Iwatsubo, T. Tomita et al., AMPA-R Removal Underlies A?-induced Synaptic Depression and Dendritic Spine Loss, Neuron, vol.52, pp.831-843, 2006.

H. Huang and Z. Jiang, Accumulated Amyloid-? Peptide and Hyperphosphorylated Tau Protein: Relationship and Links in Alzheimer's Disease, Journal of Alzheimer's Disease, vol.16, pp.15-27, 2009.

Y. Huang, C. Yang, C. Huang, M. Tai, and K. Hsu, Pharmacological and genetic accumulation of hypoxia-inducible factor-1alpha enhances excitatory synaptic transmission in hippocampal neurons through the production of vascular endothelial growth factor, J. Neurosci, vol.30, pp.6080-6093, 2010.

A. Hudmon and H. Schulman, Neuronal CA2+/calmodulin-dependent protein kinase II: the role of structure and autoregulation in cellular function, Annu. Rev. Biochem, vol.71, pp.473-510, 2002.

J. E. Huettner, Kainate receptors and synaptic transmission, Progress in Neurobiology, vol.70, pp.387-407, 2003.

B. Hyman, N. Vasdev, and R. Sperling, Tau positron emission tomographic imaging in aging and early Alzheimer disease, Ann. Neurol, vol.79, pp.110-119, 2016.

M. Iizuka, M. Abe, K. Shiiba, I. Sasaki, and Y. Sato, Down syndrome candidate region 1, a downstream target of VEGF, participates in endothelial cell migration and angiogenesis, J. Vasc. Res, vol.41, pp.334-344, 2004.

M. Ingelsson, H. Fukumoto, K. L. Newell, J. H. Growdon, E. T. Hedley-whyte et al., Early Abeta accumulation and progressive synaptic loss, gliosis, and tangle formation in AD brain, Neurology, vol.62, pp.925-931, 2004.

J. S. Isaacson and R. A. Nicoll, The uptake inhibitor L-trans-PDC enhances responses to glutamate but fails to alter the kinetics of excitatory synaptic currents in the hippocampus, J. Neurophysiol, vol.70, pp.2187-2191, 1993.

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.

C. R. Jack, D. S. Knopman, W. J. Jagust, L. M. Shaw, P. S. Aisen et al., Hypothetical model of dynamic biomarkers of the Alzheimer's pathological cascade, Lancet Neurol, vol.9, p.119, 2010.

C. E. Jahr and C. F. Stevens, Calcium permeability of the N-methyl-D-aspartate receptor channel in hippocampal neurons in culture, Proc Natl Acad Sci U S A, vol.90, pp.11573-11577, 1993.

L. B. Jakeman, M. Armanini, H. S. Phillips, and N. Ferrara, Developmental expression of binding sites and messenger ribonucleic acid for vascular endothelial growth factor suggests a role for this protein in vasculogenesis and angiogenesis, Endocrinology, vol.133, pp.848-859, 1993.

L. Jakobsson, J. Kreuger, K. Holmborn, L. Lundin, I. Eriksson et al., Heparan sulfate in trans potentiates VEGFR-mediated angiogenesis, Dev. Cell, vol.10, pp.625-634, 2006.

H. H. Jarosz-griffiths, E. Noble, J. V. Rushworth, and N. M. Hooper, Amyloid-? Receptors: The Good, the Bad, and the Prion Protein, J Biol Chem, vol.291, pp.3174-3183, 2016.

M. Jeltsch, A. Kaipainen, V. Joukov, X. Meng, M. Lakso et al., Hyperplasia of lymphatic vessels in VEGF-C transgenic mice, Science, vol.276, pp.1423-1425, 1997.

Q. S. Ji, G. E. Winnier, K. D. Niswender, D. Horstman, R. Wisdom et al., Essential role of the tyrosine kinase substrate phospholipase Cgamma1 in mammalian growth and development, Proc. Natl. Acad. Sci. U.S.A, vol.94, pp.2999-3003, 1997.

H. Jia, S. Jezequel, M. Löhr, S. Shaikh, D. Davis et al., Peptides encoded by exon 6 of VEGF inhibit endothelial cell biological responses and angiogenesis induced by VEGF, Biochem. Biophys. Res. Commun, vol.283, pp.164-173, 2001.

X. Jiang and X. Wang, Cytochrome c promotes caspase-9 activation by inducing nucleotide binding to Apaf-1, J. Biol. Chem, vol.275, pp.31199-31203, 2000.

K. Jin, Y. Zhu, Y. Sun, X. O. Mao, L. Xie et al., Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo, Proceedings of the National Academy of Sciences, vol.99, pp.11946-11950, 2002.

M. Jinnin, D. Medici, L. Park, N. Limaye, Y. Liu et al., Suppressed NFAT-dependent VEGFR1 expression and constitutive VEGFR2 signaling in infantile hemangioma, Nat Med, vol.14, pp.1236-1246, 2008.

M. Jinnin, D. Medici, L. Park, N. Limaye, Y. Liu et al., Suppressed NFAT-dependent VEGFR1 expression and constitutive VEGFR2 signaling in infantile hemangioma, Nat. Med, vol.14, pp.1236-1246, 2008.

J. Jo, D. J. Whitcomb, K. M. Olsen, T. L. Kerrigan, S. Lo et al., A?1-42 inhibition of LTP is mediated by a signaling pathway involving caspase-3, Akt1 and GSK-3?, Nature Neuroscience, vol.14, pp.545-547, 2011.

K. A. Johnson, A. Schultz, R. A. Betensky, J. A. Becker, J. Sepulcre et al., Tau positron emission tomographic imaging in aging and early Alzheimer disease, vol.79, pp.110-119, 2016.

E. G. Jones and T. P. Powell, Morphological Variations in the Dendritic Spines of the Neocortex, Journal of Cell Science, vol.5, pp.509-529, 1969.

T. L. Jones and B. T. Hyman, Amyloid accelerates tau propagation and toxicity in a model of early Alzheimer's disease, Acta Neuropathol Commun, vol.3, p.14, 2015.

W. Ju, W. Morishita, J. Tsui, G. Gaietta, T. J. Deerinck et al., Activity-dependent regulation of dendritic synthesis and trafficking of AMPA receptors, Nature Neuroscience, vol.7, pp.244-253, 2004.

M. Jucker and L. C. Walker, Self-propagation of pathogenic protein aggregates in neurodegenerative diseases, Nature, vol.501, pp.45-51, 2013.

S. Jurado, V. Biou, and R. C. Malenka, A calcineurin/AKAP complex is required for NMDA receptor-dependent long-term depression, Nature Neuroscience, vol.13, pp.1053-1055, 2010.

M. E. Kabir and J. G. Safar, Implications of prion adaptation and evolution paradigm for human neurodegenerative diseases, Prion, vol.8, pp.111-116, 2014.

M. D. Kane, W. J. Lipinski, M. J. Callahan, F. Bian, R. A. Durham et al., Evidence for Seeding of ?-Amyloid by Intracerebral Infusion of Alzheimer Brain Extracts in ?-Amyloid Precursor Protein-Transgenic Mice, The Journal of Neuroscience, vol.20, pp.3606-3611, 2000.

H. Kasai, M. Fukuda, S. Watanabe, A. Hayashi-takagi, and J. Noguchi, Structural dynamics of dendritic spines in memory and cognition, Trends in Neurosciences, vol.33, pp.121-129, 2010.

H. Kasai, M. Matsuzaki, J. Noguchi, N. Yasumatsu, and H. Nakahara, Structurestability-function relationships of dendritic spines, Trends Neurosci, vol.26, pp.360-368, 2003.

B. Katz and R. Miledi, A study of synaptic transmission in the absence of nerve impulses, The Journal of Physiology, vol.192, p.407, 1967.

L. C. Katz and C. J. Shatz, Synaptic activity and the construction of cortical circuits, Science, vol.274, pp.1133-1138, 1996.

R. Kayed, E. Head, F. Sarsoza, T. Saing, C. W. Cotman et al., Fibril specific, conformation dependent antibodies recognize a generic epitope common to amyloid fibrils and fibrillar oligomers that is absent in prefibrillar oligomers, Mol Neurodegener, vol.2, p.18, 2007.

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. L. Kendall, G. Wang, J. Disalvo, and K. A. Thomas, Specificity of vascular endothelial cell growth factor receptor ligand binding domains, Biochem. Biophys. Res. Commun, vol.201, pp.326-330, 1994.

R. L. Kendall, R. Z. Rutledge, X. Mao, A. J. Tebben, R. W. Hungate et al., , 1999.

, Vascular Endothelial Growth Factor Receptor KDR Tyrosine Kinase Activity Is Increased by Autophosphorylation of Two Activation Loop Tyrosine Residues, J. Biol. Chem, vol.274, pp.6453-6460

G. A. Kerchner and R. A. Nicoll, Silent synapses and the emergence of a postsynaptic mechanism for LTP, Nature Reviews Neuroscience, vol.9, pp.813-825, 2008.

G. A. Kerchner, G. Wang, C. Qiu, J. E. Huettner, and M. Zhuo, Direct Presynaptic Regulation of GABA/Glycine Release by Kainate Receptors in the Dorsal Horn: An Ionotropic Mechanism, Neuron, vol.32, pp.477-488, 2001.

S. S. Khan, M. Lacroix, G. Boyle, M. A. Sherman, J. L. Brown et al., Bidirectional modulation of Alzheimer phenotype by alpha-synuclein in mice and primary neurons, Acta Neuropathologica, vol.136, pp.589-605, 2018.

B. W. Kim, M. Choi, Y. Kim, H. Park, H. Lee et al., Vascular endothelial growth factor (VEGF) signaling regulates hippocampal neurons by elevation of intracellular calcium and activation of calcium/calmodulin protein kinase II and mammalian target of rapamycin, Cell. Signal, vol.20, pp.714-725, 2008.

D. Kim and Y. S. Moon, Vascular endothelial growth factor in alzheimer's disease with depressive symptoms, The Journal of the Alzheimer's Association, vol.10, pp.670-671, 2014.

H. Kim, J. Choi, J. Cha, J. Choi, and M. Lee, Expression of vascular endothelial growth factor receptors Flt-1 and Flk-1 in embryonic rat forebrain, Neuroscience Letters, vol.425, pp.131-135, 2007.

J. Kim, J. M. Basak, and D. M. Holtzman, The Role of Apolipoprotein E in Alzheimer's Disease, Neuron, vol.63, pp.287-303, 2009.

L. C. Kim, L. Song, and E. B. Haura, Src kinases as therapeutic targets for cancer, Nature Reviews Clinical Oncology, vol.6, p.587, 2009.

N. H. Kim, J. H. Oh, J. A. Seo, K. W. Lee, S. G. Kim et al., Vascular endothelial growth factor (VEGF) and soluble VEGF receptor FLT-1 in diabetic nephropathy, Kidney International, vol.67, pp.167-177, 2005.

S. Kim and E. B. Ziff, Calcineurin Mediates Synaptic Scaling Via Synaptic Trafficking of Ca2+-Permeable AMPA Receptors, PLOS Biology, vol.12, 2014.

Y. Kim, Y. Lee, M. Seo, S. Kim, J. Lee et al., Calcineurin dephosphorylates glycogen synthase kinase-3 beta at serine-9 in neuroblast-derived cells, J. Neurochem, vol.111, pp.344-354, 2009.

K. A. Kittelberger, F. Piazza, G. Tesco, and L. G. Reijmers, Natural Amyloid-Beta Oligomers Acutely Impair the Formation of a Contextual Fear Memory in Mice, PLoS ONE, vol.7, p.29940, 2012.

C. Klein, E. M. Kramer, A. M. Cardine, B. Schraven, R. Brandt et al., Process outgrowth of oligodendrocytes is promoted by interaction of fyn kinase with the cytoskeletal protein tau, J. Neurosci, vol.22, issue.3, pp.698-707, 2002.

A. M. Kleschevnikov and A. Routtenberg, PKC activation rescues LTP from NMDA receptor blockade, Hippocampus, vol.11, pp.168-175, 2001.

S. Koch and L. Claesson-welsh, Signal Transduction by Vascular Endothelial Growth Factor Receptors. Cold Spring Harb Perspect Med, vol.2, 2012.

S. Koch, S. Tugues, X. Li, L. Gualandi, C. et al., Signal transduction by vascular endothelial growth factor receptors, Biochemical Journal, vol.437, pp.169-183, 2011.

R. M. Koffie, M. Meyer-luehmann, T. Hashimoto, K. W. Adams, M. L. Mielke et al., Oligomeric amyloid beta associates with postsynaptic densities and correlates with excitatory synapse loss near senile plaques, vol.106, pp.4012-4017, 2009.

K. Koltowska, S. Paterson, N. I. Bower, G. J. Baillie, A. K. Lagendijk et al., mafba is a downstream transcriptional effector of Vegfc signaling essential for embryonic lymphangiogenesis in zebrafish, Genes Dev, vol.29, pp.1618-1630, 2015.

U. Konietzko, AICD Nuclear Signaling and Its Possible Contribution to Alzheimers Disease, Current Alzheimer Research, vol.9, pp.200-216, 2012.

C. D. Kopec, B. Li, W. Wei, J. Boehm, and R. Malinow, Glutamate receptor exocytosis and spine enlargement during chemically induced long-term potentiation, J. Neurosci, vol.26, 2000.

E. Krämer-albers and R. White, From axon-glial signalling to myelination: the integrating role of oligodendroglial Fyn kinase, Cellular and Molecular Life Sciences, vol.68, 2003.

J. J. Kril, S. Patel, A. J. Harding, and G. M. Halliday, Neuron loss from the hippocampus of Alzheimer's disease exceeds extracellular neurofibrillary tangle formation, Acta Neuropathol, vol.103, pp.370-376, 2002.

J. J. Krupp, B. Vissel, C. G. Thomas, S. F. Heinemann, and G. L. Westbrook, Calcineurin acts via the C-terminus of NR2A to modulate desensitization of NMDA receptors, Neuropharmacology, vol.42, pp.593-602, 2002.

S. Kundi, R. Bicknell, and Z. Ahmed, The role of angiogenic and wound-healing factors after spinal cord injury in mammals, Neuroscience Research, vol.76, pp.1-9, 2013.

L. Kung, T. Batiuk, S. Palomo-pinon, J. Noujaim, L. M. Helms et al., Tissue distribution of calcineurin and its sensitivity to inhibition by cyclosporine, Am J Transplant, vol.1, issue.4, pp.325-333, 2001.

R. Kuruvilla, L. S. Zweifel, N. O. Glebova, B. E. Lonze, G. Valdez et al., A Neurotrophin Signaling Cascade Coordinates Sympathetic Neuron Development through Differential Control of TrkA Trafficking and Retrograde Signaling, Cell, vol.118, pp.243-255, 2004.

L. Labrecque, I. Royal, D. S. Surprenant, C. Patterson, D. Gingras et al., Regulation of Vascular Endothelial Growth Factor Receptor-2 Activity by Caveolin-1 and Plasma Membrane Cholesterol, Mol Biol Cell, vol.14, pp.334-347, 2003.

P. N. Lacor, M. C. Buniel, L. Chang, S. J. Fernandez, Y. Gong et al., Synaptic targeting by Alzheimer's-related amyloid beta oligomers, J. Neurosci, vol.24, pp.10191-10200, 2004.

P. N. Lacor, M. C. Buniel, P. W. Furlow, A. S. Clemente, P. T. Velasco et al., A? Oligomer-Induced Aberrations in Synapse Composition, Shape, and Density Provide a Molecular Basis for Loss of Connectivity in Alzheimer's Disease, J. Neurosci, vol.27, pp.796-807, 2007.

P. N. Lacor, M. C. Buniel, P. W. Furlow, A. S. Clemente, P. T. Velasco et al., Abeta oligomer-induced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer's disease, J. Neurosci, vol.27, pp.796-807, 2007.

F. M. Laferla, K. N. Green, and S. Oddo, Intracellular amyloid-? in Alzheimer's disease, Nature Reviews Neuroscience, vol.8, pp.499-509, 2007.

J. E. Lähteenvuo, M. T. Lähteenvuo, A. Kivelä, C. Rosenlew, A. Falkevall et al., Vascular endothelial growth factor-B induces myocardium-specific angiogenesis and arteriogenesis via vascular endothelial growth factor receptor-1-and neuropilin receptor-1-dependent mechanisms, Circulation, vol.119, pp.845-856, 2009.

B. K. Lal, S. Varma, P. J. Pappas, R. W. Hobson, and W. N. Durán, VEGF increases permeability of the endothelial cell monolayer by activation of PKB/akt, endothelial nitric-oxide synthase, and MAP kinase pathways, Microvasc. Res, vol.62, pp.252-262, 2001.

L. Lamalice, F. Houle, and J. Huot, Phosphorylation of Tyr1214 within VEGFR-2 triggers the recruitment of Nck and activation of Fyn leading to SAPK2/p38 activation and endothelial cell migration in response to VEGF, J. Biol. Chem, vol.281, pp.34009-34020, 2006.

M. G. Lampugnani, F. Orsenigo, M. C. Gagliani, C. Tacchetti, and E. Dejana, Vascular endothelial cadherin controls VEGFR-2 internalization and signaling from intracellular compartments, J Cell Biol, vol.174, pp.593-604, 2006.

D. M. Landis and T. S. Reese, Cytoplasmic organization in cerebellar dendritic spines, J. Cell Biol, vol.97, pp.1169-1178, 1983.

M. Larson, M. A. Sherman, F. Amar, M. Nuvolone, J. A. Schneider et al., The Complex PrPc-Fyn Couples Human Oligomeric A with Pathological Tau Changes in Alzheimer's Disease, Journal of Neuroscience, vol.32, pp.16857-16871, 2012.

M. E. Larson, S. J. Greimel, F. Amar, M. Lacroix, G. Boyle et al., Selective lowering of synapsins induced by oligomeric ?-synuclein exacerbates memory deficits, vol.114, pp.4648-4657, 2017.

M. E. Larson and S. E. Lesné, Soluble A? oligomer production and toxicity: Soluble oligomeric A? production and toxicity, Journal of Neurochemistry, vol.120, pp.125-139, 2012.

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.

S. E. Lauri, Z. A. Bortolotto, D. Bleakman, P. L. Ornstein, D. Lodge et al., A critical role of a facilitatory presynaptic kainate receptor in mossy fiber LTP, Neuron, vol.32, pp.697-709, 2001.

I. N. Lavrik, A. Golks, and P. H. Krammer, Caspases: pharmacological manipulation of cell death, J Clin Invest, vol.115, pp.2665-2672, 2005.

H. Lee, M. Barbarosie, K. Kameyama, M. F. Bear, and R. L. Huganir, Regulation of distinct AMPA receptor phosphorylation sites during bidirectional synaptic plasticity, Nature, vol.405, pp.955-959, 2000.

H. Lee, K. Kameyama, R. L. Huganir, and M. F. Bear, NMDA Induces Long-Term Synaptic Depression and Dephosphorylation of the GluR1 Subunit of AMPA Receptors in Hippocampus, Neuron, vol.21, pp.1151-1162, 1998.

H. Lee, K. Takamiya, J. Han, H. Man, C. Kim et al., Phosphorylation of the AMPA Receptor GluR1 Subunit Is Required for Synaptic Plasticity and Retention of Spatial Memory, Cell, vol.112, pp.631-643, 2003.

H. Lee, K. Takamiya, K. He, L. Song, and R. L. Huganir, Specific Roles of AMPA Receptor Subunit GluR1 (GluA1) Phosphorylation Sites in Regulating Synaptic Plasticity in the CA1 Region of Hippocampus, Journal of Neurophysiology, vol.103, pp.479-489, 2010.

T. Lee, J. Folkman, and K. Javaherian, HSPG-binding peptide corresponding to the exon 6a-encoded domain of VEGF inhibits tumor growth by blocking angiogenesis in murine model, PLoS ONE, vol.5, p.9945, 2010.

M. A. Lemmon and J. Schlessinger, Cell signaling by receptor tyrosine kinases, Cell, vol.141, pp.1117-1134, 2010.

A. S. Leonard, M. A. Davare, M. C. Horne, C. C. Garner, and J. W. Hell, SAP97 Is Associated with the Amino-3-hydroxy-5-methylisoxazole-4-propionic Acid Receptor GluR1 Subunit, J. Biol. Chem, p.31, 1998.

J. R. Leonard, B. J. Klocke, C. Sa, R. A. Flavell, and K. A. Roth, Strain-dependent neurodevelopmental abnormalities in caspase-3-deficient mice, J. Neuropathol. Exp. Neurol, vol.61, pp.673-677, 2002.

S. Lesné, M. T. Koh, L. Kotilinek, R. Kayed, C. G. Glabe et al., A specific amyloid-? protein assembly in the brain impairs memory, Nature, vol.440, pp.352-357, 2006.

S. E. Lesné, Toxic oligomer species of amyloid-? in Alzheimer's disease, a timing issue, Swiss Med Wkly, vol.144, p.14021, 2014.

S. E. Lesné, M. A. Sherman, M. Grant, M. Kuskowski, J. A. Schneider et al., Brain amyloid-? oligomers in ageing and Alzheimer's disease, Brain, vol.136, pp.1383-1398, 2013.

Y. Y. Leung, J. B. Toledo, A. Nefedov, R. Polikar, N. Raghavan et al., Identifying amyloid pathology-related cerebrospinal fluid biomarkers for Alzheimer's disease in a multicohort study, Alzheimer's & Dementia: Diagnosis, Assessment & Disease Monitoring, vol.1, pp.339-348, 2015.

E. Levy, M. D. Carman, I. J. Fernandez-madrid, M. D. Power, I. Lieberburg et al., Mutation of the Alzheimer's disease amyloid gene in hereditary cerebral hemorrhage, Dutch type, Science, vol.248, pp.1124-1126, 1990.

N. Li, K. Liu, Y. Qiu, Z. Ren, R. Dai et al., Effect of Presenilin Mutations on APP Cleavage; Insights into the Pathogenesis of FAD, Front Aging Neurosci, vol.8, 2016.

S. Li, S. Hong, N. E. Shepardson, D. M. Walsh, G. M. Shankar et al., Soluble Oligomers of Amyloid ? Protein Facilitate Hippocampal Long-Term Depression by Disrupting Neuronal Glutamate Uptake, Neuron, vol.62, pp.788-801, 2009.

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, vol.31, pp.6627-6638, 2011.

X. Li, C. Lee, Z. Tang, F. Zhang, P. Arjunan et al., VEGF-B: a survival, or an angiogenic factor?, Cell Adh Migr, vol.3, pp.322-327, 2009.

X. Li, N. Padhan, E. O. Sjöström, F. P. Roche, C. Testini et al., VEGFR2 pY949 signalling regulates adherens junction integrity and metastatic spread, vol.7, p.11017, 2016.

X. Li, M. Tjwa, I. Van-hove, B. Enholm, E. Neven et al., Reevaluation of the role of VEGF-B suggests a restricted role in the revascularization of the ischemic myocardium, Arterioscler. Thromb. Vasc. Biol, vol.28, pp.1614-1620, 2008.

Z. Li, J. Jo, J. Jia, S. Lo, D. J. Whitcomb et al., Caspase-3 activation via mitochondria is required for long-term depression and AMPA receptor internalization, Cell, vol.141, pp.859-871, 2010.

Q. Lian, C. J. Ladner, D. Magnuson, and J. M. Lee, Selective changes of calcineurin (protein phosphatase 2B) activity in Alzheimer's disease cerebral cortex, Exp. Neurol, vol.167, pp.158-165, 2001.

H. Liao, T. Kume, C. Mckay, M. Xu, J. N. Ihle et al., Absence of erythrogenesis and vasculogenesis in Plcg1-deficient mice, J. Biol. Chem, vol.277, pp.9335-9341, 2002.

T. Licht, R. Eavri, I. Goshen, Y. Shlomai, A. Mizrahi et al., VEGF is required for dendritogenesis of newly born olfactory bulb interneurons, Development, vol.137, pp.261-271, 2010.

T. Licht, I. Goshen, A. Avital, T. Kreisel, S. Zubedat et al., Reversible modulations of neuronal plasticity by VEGF, vol.108, pp.5081-5086, 2011.

D. N. Lieberman and I. Mody, Regulation of NMDA channel function by endogenous Ca2+-dependent phosphatase, Nature, vol.369, pp.235-239, 1994.

J. Lisman, A mechanism for the Hebb and the anti-Hebb processes underlying learning and memory, Proceedings of the National Academy of Sciences, vol.86, pp.9574-9578, 1989.

J. Lisman, H. Schulman, and H. Cline, The molecular basis of CaMKII function in synaptic and behavioural memory, Nat. Rev. Neurosci, vol.3, pp.175-190, 2002.

L. Liu, T. P. Wong, M. F. Pozza, K. Lingenhoehl, Y. Wang et al., Role of NMDA Receptor Subtypes in Governing the Direction of Hippocampal Synaptic Plasticity, Science, vol.304, pp.1021-1024, 2004.

N. Louneva, J. W. Cohen, L. Han, K. Talbot, R. S. Wilson et al., Caspase-3 Is Enriched in Postsynaptic Densities and Increased in Alzheimer's Disease, Am J Pathol, vol.173, pp.1488-1495, 2008.

Y. Lu, K. Tomizawa, A. Moriwaki, Y. Hayashi, M. Tokuda et al., Calcineurin inhibitors, FK506 and cyclosporin A, suppress the NMDA receptor-mediated potentials and LTP, but not depotentiation in the rat hippocampus, Brain Research, vol.729, pp.142-146, 1996.

Y. M. Lu, J. C. Roder, J. Davidow, and M. W. Salter, Src Activation in the Induction of Long-Term Potentiation in CAI Hippocampal Neurons, p.5, 1998.

C. Lüscher and R. C. Malenka, NMDA Receptor-Dependent Long-Term Potentiation and Long-Term Depression (LTP/LTD), Cold Spring Harb Perspect Biol, vol.4, p.5710, 2012.

M. P. Lussier, A. Sanz-clemente, and K. W. Roche, Dynamic Regulation of N -Methyl-daspartate (NMDA) and ?-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA) Receptors by Posttranslational Modifications, Journal of Biological Chemistry, vol.290, pp.28596-28603, 2015.

G. Lynch, J. Larson, S. Kelso, G. Barrionuevo, and F. Schottler, Intracellular injections of EGTA block induction of hippocampal long-term potentiation, Nature, vol.305, pp.719-721, 1983.

F. Mac-gabhann and A. S. Popel, Dimerization of VEGF receptors and implications for signal transduction: a computational study, Biophys Chem, vol.128, pp.125-139, 2007.

F. Mackenzie and C. Ruhrberg, Diverse roles for VEGF-A in the nervous system, Development, vol.139, pp.1371-1380, 2012.

S. D. Madden and T. G. Cotter, Cell Death in Brain Development and Degeneration: Control of Caspase Expression May Be Key! Molecular Neurobiology, vol.37, pp.1-6, 2008.

A. S. Maharaj, M. Saint-geniez, A. E. Maldonado, and P. A. D'amore, Vascular Endothelial Growth Factor Localization in the Adult, The American Journal of Pathology, vol.168, p.639, 2006.

R. R. Mahmmoud, S. Sase, Y. D. Aher, A. Sase, M. Gröger et al., Spatial and Working Memory Is Linked to Spine Density and Mushroom Spines, PLOS ONE, vol.10, p.139739, 2015.

R. C. Malenka, Synaptic plasticity in the hippocampus: LTP and LTD, Cell, vol.78, pp.535-538, 1994.

R. C. Malenka and M. F. Bear, LTP and LTD: An Embarrassment of Riches, Neuron, vol.44, pp.5-21, 2004.

R. Malinow and R. C. Malenka, AMPA Receptor Trafficking and Synaptic Plasticity, Annual Review of Neuroscience, vol.25, pp.103-126, 2002.

G. Malleret, J. M. Alarcon, G. Martel, S. Takizawa, S. Vronskaya et al., Bidirectional Regulation of Hippocampal Long-Term Synaptic Plasticity and Its Influence on Opposing Forms of Memory, J. Neurosci, vol.30, pp.3813-3825, 2010.

S. J. Mandriota, G. Seghezzi, J. Vassalli, N. Ferrara, S. Wasi et al., Vascular Endothelial Growth Factor Increases Urokinase Receptor Expression in Vascular Endothelial Cells, J. Biol. Chem, vol.270, pp.9709-9716, 1995.

M. Maravall, I. Y. Koh, W. B. Lindquist, and K. Svoboda, Experience-dependent changes in basal dendritic branching of layer 2/3 pyramidal neurons during a critical period for developmental plasticity in rat barrel cortex, Cereb. Cortex, vol.14, pp.655-664, 2004.

G. S. Marco, S. Reuter, U. Hillebrand, S. Amler, M. König et al., The Soluble VEGF Receptor sFlt1 Contributes to Endothelial Dysfunction in CKD, J Am Soc Nephrol, vol.20, pp.2235-2245, 2009.

S. Markovic-mueller, E. Stuttfeld, M. Asthana, T. Weinert, S. Bliven et al., Structure of the Full-length VEGFR-1, 2017.

, Extracellular Domain in Complex with VEGF-A. Structure, vol.25, pp.341-352

E. Masliah, R. D. Terry, M. Mallory, M. Alford, and L. A. Hansen, Diffuse plaques do not accentuate synapse loss in Alzheimer's disease, Am. J. Pathol, vol.137, pp.1293-1297, 1990.

I. Mateo, J. Llorca, J. Infante, E. Rodríguez-rodríguez, C. Fernández-viadero et al., Low serum VEGF levels are associated with Alzheimer's disease, Acta Neurol. Scand, vol.116, pp.56-58, 2007.

N. Matsuo, L. Reijmers, and M. Mayford, Spine-Type-Specific Recruitment of Newly Synthesized AMPA Receptors with Learning, Science, vol.319, pp.1104-1107, 2008.

M. Matsuzaki, N. Honkura, G. C. Ellis-davies, and H. Kasai, Structural basis of longterm potentiation in single dendritic spines, Nature, vol.429, pp.761-766, 2004.

C. M. Mazure and J. Swendsen, Sex differences in Alzheimer's disease and other dementias, Lancet Neurol, vol.15, pp.451-452, 2016.

D. P. Mccloskey, S. D. Croll, and H. E. Scharfman, Depression of Synaptic Transmission by Vascular Endothelial Growth Factor in Adult Rat Hippocampus and Evidence for Increased Efficacy after Chronic Seizures, The Journal of neuroscience : the official journal of the Society for Neuroscience, vol.25, p.8889, 2005.

V. Mecollari, B. Nieuwenhuis, and J. Verhaagen, A perspective on the role of class III semaphorin signaling in central nervous system trauma, Front. Cell. Neurosci, vol.8, 2014.

C. Meissirel, C. R. Almodovar, . De, E. Knevels, C. Coulon et al., VEGF modulates NMDA receptors activity in cerebellar granule cells through Src-family kinases before synapse formation, vol.108, pp.13782-13787, 2011.

D. Meyer, T. Bonhoeffer, and V. Scheuss, Balance and Stability of Synaptic Structures during Synaptic Plasticity, Neuron, vol.82, pp.430-443, 2014.

E. C. Miller, P. J. Teravskis, B. W. Dummer, X. Zhao, R. L. Huganir et al., Tau phosphorylation and tau mislocalization mediate soluble A? oligomer-induced AMPA glutamate receptor signaling deficits, Eur J Neurosci, vol.39, pp.1214-1224, 2014.

M. Miller and A. Peters, Maturation of rat visual cortex. II. A combined Golgi-electron microscope study of pyramidal neurons, J. Comp. Neurol, vol.203, pp.555-573, 1981.

A. Minchenko, T. Bauer, S. Salceda, and J. Caro, Hypoxic stimulation of vascular endothelial growth factor expression in vitro and in vivo, Lab. Invest, vol.71, pp.374-379, 1994.

M. R. Minter, J. M. Taylor, and P. J. Crack, The contribution of neuroinflammation to amyloid toxicity in Alzheimer's disease, Journal of Neurochemistry, vol.136, pp.457-474, 2016.

T. Miyakawa, L. M. Leiter, D. J. Gerber, R. R. Gainetdinov, T. D. Sotnikova et al., Conditional calcineurin knockout mice exhibit multiple abnormal behaviors related to schizophrenia, PNAS, vol.100, pp.8987-8992, 2003.

T. Miyake, K. Kumasawa, N. Sato, T. Takiuchi, H. Nakamura et al., Soluble VEGF receptor 1 (sFLT1) induces non-apoptotic death in ovarian and colorectal cancer cells, Sci Rep, vol.6, p.24853, 2016.

T. Moehlmann, E. Winkler, X. Xia, D. Edbauer, J. Murrell et al., Presenilin-1 mutations of leucine 166 equally affect the generation of the Notch and APP intracellular domains independent of their effect on Abeta 42 production, Proc. Natl. Acad. Sci. U.S.A, vol.99, pp.8025-8030, 2002.

H. Monyer, N. Burnashev, D. J. Laurie, B. Sakmann, and P. H. Seeburg, Developmental and regional expression in the rat brain and functional properties of four NMDA receptors, Neuron, vol.12, pp.529-540, 1994.

R. G. Morris, E. Anderson, G. S. Lynch, and M. Baudry, Selective impairment of learning and blockade of long-term potentiation by an N-methyl-D-aspartate receptor antagonist, AP5, Nature, p.774, 1986.

J. Mothet, A. T. Parent, H. Wolosker, R. O. Brady, D. J. Linden et al., d-Serine is an endogenous ligand for the glycine site of the N-methyl-d-aspartate receptor, vol.97, pp.4926-4931, 2000.

R. Motter, C. Vigo-pelfrey, D. Kholodenko, R. Barbour, K. Johnson-wood et al., Reduction of beta-amyloid peptide42 in the cerebrospinal fluid of patients with Alzheimer's disease, Ann. Neurol, vol.38, pp.643-648, 1995.

D. E. Moustaine, S. Granier, E. Doumazane, P. Scholler, R. Rahmeh et al., Distinct roles of metabotropic glutamate receptor dimerization in agonist activation and G-protein coupling, vol.109, pp.16342-16347, 2012.

L. Mucke and D. J. Selkoe, Neurotoxicity of Amyloid -Protein: Synaptic and Network Dysfunction. Cold Spring Harbor Perspectives in Medicine, vol.2, pp.6338-006338, 2012.

N. Mukerjee, K. M. Mcginnis, M. E. Gnegy, and K. K. Wang, Caspase-Mediated Calcineurin Activation Contributes to IL-2 Release during, T Cell Activation. Biochemical and Biophysical Research Communications, vol.285, pp.1192-1199, 2001.

Y. Mukouyama, D. Shin, S. Britsch, M. Taniguchi, and D. J. Anderson, Sensory Nerves Determine the Pattern of Arterial Differentiation and Blood Vessel Branching in the Skin, Cell, vol.109, pp.693-705, 2002.

R. M. Mulkey, S. Endo, S. Shenolikar, and R. C. Malenka, Involvement of a calcineurin/inhibitor-1 phosphatase cascade in hippocampal long-term depression, Nature, vol.369, pp.486-488, 1994.

Y. A. Muller, H. W. Christinger, B. A. Keyt, and A. M. De-vos, The crystal structure of vascular endothelial growth factor (VEGF) refined to 1.93 Å resolution: multiple copy flexibility and receptor binding, Structure, vol.5, pp.1325-1338, 1997.

Y. A. Muller, C. Heiring, R. Misselwitz, K. Welfle, and H. Welfle, The Cystine Knot Promotes Folding and Not Thermodynamic Stability in Vascular Endothelial Growth Factor, J. Biol. Chem, vol.277, pp.43410-43416, 2002.

E. M. Muñoz and R. J. Linhardt, Heparin-Binding Domains in Vascular Biology, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.24, pp.1549-1557, 2004.

M. P. Murphy and H. Levine, Alzheimer's Disease and the ?-Amyloid Peptide, J Alzheimers Dis, vol.19, p.311, 2010.

E. S. Musiek and D. M. Holtzman, Three Dimensions of the Amyloid Hypothesis: Time, Space, and "Wingmen, vol.18, pp.800-806, 2015.

S. Nada, T. Yagi, H. Takeda, T. Tokunaga, H. Nakagawa et al., Constitutive activation of Src family kinases in mouse embryos that lack Csk, Cell, vol.73, pp.1125-1135, 1993.

U. V. Nägerl, N. Eberhorn, S. B. Cambridge, and T. Bonhoeffer, Bidirectional activitydependent morphological plasticity in hippocampal neurons, Neuron, vol.44, pp.759-767, 2004.

T. Nakazawa, S. Komai, T. Tezuka, C. Hisatsune, H. Umemori et al., Characterization of Fyn-mediated Tyrosine Phosphorylation Sites on GluR?2 (NR2B) Subunit of theN-Methyl-d-aspartate Receptor, J. Biol. Chem, vol.276, pp.693-699, 2001.

A. Nasir, T. R. Holzer, M. Chen, M. Z. Man, and A. E. Schade, Differential expression of VEGFR2 protein in HER2 positive primary human breast cancer: potential relevance to antiangiogenic therapies, Cancer Cell Int, vol.17, 2017.

Z. S. Nasreddine, N. A. Phillips, V. Bédirian, S. Charbonneau, V. Whitehead et al., The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment, J Am Geriatr Soc, vol.53, pp.695-699, 2005.

R. E. Nicholls, J. M. Alarcon, G. Malleret, R. C. Carroll, M. Grody et al., Transgenic mice lacking NMDAR-dependent LTD exhibit deficits in behavioral flexibility, Neuron, vol.58, pp.104-117, 2008.

O. Nicole, S. Hadzibegovic, J. Gajda, B. Bontempi, T. Bem et al., Soluble amyloid beta oligomers block the learning-induced increase in hippocampal sharp wave-ripple rate and impair spatial memory formation, Scientific Reports, p.6, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01284761

F. Nicoletti, J. Bockaert, G. L. Collingridge, P. J. Conn, F. Ferraguti et al., Metabotropic glutamate receptors: from the workbench to the bedside, Neuropharmacology, vol.60, pp.1017-1041, 2011.

C. Nilsberth, A. Westlind-danielsson, C. B. Eckman, M. M. Condron, K. Axelman et al., The "Arctic" APP mutation (E693G) causes Alzheimer's disease by enhanced Abeta protofibril formation, Nat. Neurosci, vol.4, pp.887-893, 2001.

E. A. Nimchinsky, B. L. Sabatini, and K. Svoboda, Structure and function of dendritic spines, Annu. Rev. Physiol, vol.64, pp.313-353, 2002.

C. M. Niswender and P. J. Conn, Metabotropic Glutamate Receptors: Physiology, Pharmacology, and Disease, Annual Review of Pharmacology and Toxicology, vol.50, pp.295-322, 2010.

H. Niu, I. Álvarez-Álvarez, F. Guillén-grima, and I. Aguinaga-ontoso, Prevalence and incidence of Alzheimer's disease in Europe: A meta-analysis, Neurología, english edition, vol.32, pp.523-532, 2017.

E. Nwabuisi-heath, M. J. Ladu, and C. Yu, Simultaneous analysis of dendritic spine density, morphology and glutamate receptors during neuron maturation in vitro by quantitative immunocytochemistry, J Neurosci Methods, vol.207, pp.137-147, 2012.

S. Oddo, A. Caccamo, I. F. Smith, K. N. Green, and F. M. Laferla, A Dynamic Relationship between Intracellular and Extracellular Pools of A?, Am J Pathol, vol.168, pp.184-194, 2006.

M. Oka, N. Fujisaki, A. Maruko-otake, Y. Ohtake, S. Shimizu et al., Ca2+/calmodulin-dependent protein kinase II promotes neurodegeneration caused by tau phosphorylated at Ser262/356 in a transgenic Drosophila model of tauopathy, J. Biochem, vol.162, pp.335-342, 2017.

K. Okamoto, T. Nagai, A. Miyawaki, and Y. Hayashi, Rapid and persistent modulation of actin dynamics regulates postsynaptic reorganization underlying bidirectional plasticity, Nat. Neurosci, vol.7, pp.1104-1112, 2004.

H. Okazaki, A. Hayashi-takagi, A. Nagaoka, M. Negishi, H. Ucar et al., Calcineurin knockout mice show a selective loss of small spines, Neuroscience Letters, vol.671, pp.99-102, 2018.

B. Oosthuyse, L. Moons, E. Storkebaum, H. Beck, D. Nuyens et al., Deletion of the hypoxia-response element in the vascular endothelial growth factor promoter causes motor neuron degeneration, Nature Genetics, vol.28, p.131, 2001.

S. Oray, A. Majewska, and M. Sur, Effects of synaptic activity on dendritic spine motility of developing cortical layer v pyramidal neurons, Cereb. Cortex, vol.16, pp.730-741, 2006.

R. Ossenkoppele, D. R. Schonhaut, M. Schöll, S. N. Lockhart, N. Ayakta et al., Tau PET patterns mirror clinical and neuroanatomical variability in Alzheimer's disease, vol.139, pp.1551-1567, 2016.

T. D. Palmer, A. R. Willhoite, and F. H. Gage, Vascular niche for adult hippocampal neurogenesis, Journal of Comparative Neurology, vol.425, pp.479-494, 2000.

P. Paoletti, C. Bellone, and Q. Zhou, NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease, Nature Reviews Neuroscience, vol.14, pp.383-400, 2013.

T. Papouin, L. Ladépêche, J. Ruel, S. Sacchi, M. Labasque et al., Synaptic and Extrasynaptic NMDA Receptors Are Gated by Different Endogenous Coagonists, Cell, vol.150, pp.633-646, 2012.

T. Papouin and S. H. Oliet, Organization, control and function of extrasynaptic NMDA receptors, Philos Trans R Soc Lond B Biol Sci, p.369, 2014.

M. Pascual-lucas, S. Viana-da-silva, M. Di-scala, C. Garcia-barroso, G. Gonzalezaseguinolaza et al., Insulin-like growth factor 2 reverses memory and synaptic deficits in APP transgenic mice, EMBO Molecular Medicine, vol.6, pp.1246-1262, 2014.

M. Passafaro, V. Piëch, and M. Sheng, Subunit-specific temporal and spatial patterns of AMPA receptor exocytosis in hippocampal neurons, Nature Neuroscience, vol.4, pp.917-926, 2001.

N. S. Patel, V. S. Mathura, C. Bachmeier, D. Beaulieu-abdelahad, V. Laporte et al., Alzheimer's beta-amyloid peptide blocks vascular endothelial growth factor mediated signaling via direct interaction with VEGFR-2, J. Neurochem, vol.112, pp.66-76, 2010.

T. Pawson, G. D. Gish, and P. Nash, SH2 domains, interaction modules and cellular wiring, Trends in Cell Biology, vol.11, pp.504-511, 2001.

A. A. Penn and C. J. Shatz, Brain Waves and Brain Wiring: The Role of Endogenous and Sensory-Driven Neural Activity in Development, Pediatric Research, vol.45, p.447, 1999.

R. G. Perez, S. Soriano, J. D. Hayes, B. Ostaszewski, W. Xia et al., Mutagenesis Identifies New Signals for ?-Amyloid Precursor Protein Endocytosis, Turnover, and the Generation of Secreted Fragments, Including A?42, J. Biol. Chem, vol.274, pp.18851-18856, 1999.

I. Pérez-otaño and M. D. Ehlers, Homeostatic plasticity and NMDA receptor trafficking, Trends in Neurosciences, vol.28, pp.229-238, 2005.

R. S. Petralia, N. Sans, Y. Wang, and R. J. Wenthold, Ontogeny of Postsynaptic Density Proteins at Glutamatergic Synapses, Mol Cell Neurosci, vol.29, pp.436-452, 2005.

H. J. Pi, N. Otmakhov, F. El-gaamouch, D. Lemelin, P. De-koninck et al., CaMKII control of spine size and synaptic strength: Role of phosphorylation states and nonenzymatic action, Proc Natl Acad Sci U S A, vol.107, pp.14437-14442, 2010.

I. Piaceri, B. Nacmias, and S. Sorbi, Genetics of familial and sporadic Alzheimer's disease. Front Biosci, vol.5, pp.167-177, 2013.

P. S. Pinheiro and C. Mulle, Presynaptic glutamate receptors: physiological functions and mechanisms of action, Nat. Rev. Neurosci, vol.9, pp.423-436, 2008.

M. M. Pleiss, P. Sompol, S. D. Kraner, H. M. Abdul, J. L. Furman et al., Calcineurin proteolysis in astrocytes: Implications for impaired synaptic function, Biochim. Biophys. Acta, vol.1862, pp.1521-1532, 2016.

M. B. Podlisny, D. T. Stephenson, M. P. Frosch, I. Lieberburg, J. A. Clemens et al., Synthetic amyloid beta-protein fails to produce specific neurotoxicity in monkey cerebral cortex, Neurobiol. Aging, vol.13, pp.561-567, 1992.

K. Poesen, D. Lambrechts, P. Van-damme, J. Dhondt, F. Bender et al., Novel role for vascular endothelial growth factor (VEGF) receptor-1 and its ligand VEGF-B in motor neuron degeneration, J. Neurosci, vol.28, pp.10451-10459, 2008.

J. Pokorný and T. Yamamoto, Postnatal ontogenesis of hippocampal CA1 area in rats. I. Development of dendritic arborisation in pyramidal neurons, Brain Res. Bull, vol.7, pp.113-120, 1981.

A. M. Pooler, M. Polydoro, E. A. Maury, S. B. Nicholls, S. M. Reddy et al., Amyloid accelerates tau propagation and toxicity in a model of early Alzheimer's disease, 2015.

E. Popugaeva, E. Pchitskaya, and I. Bezprozvanny, Dysregulation of neuronal calcium homeostasis in Alzheimer's disease -a therapeutic opportunity?, Biochem. Biophys. Res. Commun, vol.483, pp.998-1004, 2017.

P. E. Potter, P. K. Rauschkolb, Y. Pandya, L. I. Sue, M. N. Sabbagh et al., Pre-and post-synaptic cortical cholinergic deficits are proportional to amyloid plaque presence and density at preclinical stages of alzheimer's disease, Acta Neuropathol, vol.122, pp.49-60, 2011.

J. Puoliväli, J. Wang, T. Heikkinen, M. Heikkilä, T. Tapiola et al., Hippocampal A?42 Levels Correlate with Spatial Memory Deficit in APP and PS1 Double Transgenic Mice, Neurobiology of Disease, vol.9, pp.339-347, 2002.

C. Qiu, M. Kivipelto, and E. Strauss, Epidemiology of Alzheimer's disease: occurrence, determinants, and strategies toward intervention, Dialogues Clin Neurosci, vol.11, pp.111-128, 2009.

R. Radde, T. Bolmont, S. A. Kaeser, J. Coomaraswamy, D. Lindau et al., A?42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology, EMBO reports, vol.7, pp.940-946, 2006.

P. Rakic, J. P. Bourgeois, M. F. Eckenhoff, N. Zecevic, and P. S. Goldman-rakic, Concurrent overproduction of synapses in diverse regions of the primate cerebral cortex, Science, vol.232, pp.232-235, 1986.

S. Ramakrishnan, V. Anand, and S. Roy, Vascular Endothelial growth factor signaling in hypoxia and Inflammation, J Neuroimmune Pharmacol, vol.9, pp.142-160, 2014.

S. Ramón-y-cajal, Dolores del parto considerablemente atenuados por la sugestión hipnótica, Gac. Méd. Catalana (Barc.), vol.12, pp.484-486, 1889.

S. Ram?n-y-cajal, La textura del sistema nerviosa del hombre y los vertebrados, 1899.

J. Rasmussen, J. Mahler, N. Beschorner, S. A. Kaeser, L. M. Häsler et al., Amyloid polymorphisms constitute distinct clouds of conformational variants in different etiological subtypes of Alzheimer's disease, vol.114, pp.13018-13023, 2017.

M. N. Reed, J. J. Hofmeister, L. Jungbauer, A. T. Welzel, C. Yu et al., Cognitive effects of cellderived and synthetically derived A? oligomers, Neurobiology of Aging, vol.32, pp.1784-1794, 2011.

L. C. Reese, W. Zhang, K. T. Dineley, R. Kayed, and G. Taglialatela, Selective Induction Of Calcineurin Activity And Signaling By Oligomeric Amyloid Beta, Aging Cell, vol.7, pp.824-835, 2008.

W. G. Regehr and D. W. Tank, Postsynaptic NMDA receptor-mediated calcium accumulation in hippocampal CA1 pyramidal cell dendrites, Nature, vol.345, pp.807-810, 1990.

P. Religa, R. Cao, D. Religa, Y. Xue, N. Bogdanovic et al., VEGF significantly restores impaired memory behavior in Alzheimer's mice by improvement of vascular survival, Scientific Reports, 1923.

M. Renner, D. Choquet, and A. Triller, Control of the postsynaptic membrane viscosity, J. Neurosci, vol.29, pp.2926-2937, 2009.

C. Ripoli, S. Cocco, D. D. Puma, R. Piacentini, A. Mastrodonato et al., Intracellular Accumulation of Amyloid-? (A?) Protein Plays a Major Role in A?-Induced Alterations of Glutamatergic Synaptic Transmission and Plasticity, J. Neurosci, vol.34, pp.12893-12903, 2014.

W. C. Risher, T. Ustunkaya, J. S. Alvarado, and C. Eroglu, Rapid Golgi Analysis Method for Efficient and Unbiased Classification of Dendritic Spines, PLOS ONE, vol.9, p.107591, 2014.

L. Rizzi, I. Rosset, and M. Roriz-cruz, Global Epidemiology of Dementia: Alzheimer's and Vascular Types, Biomed Res Int, vol.2014, 2014.

K. W. Roche, R. J. O'brien, A. L. Mammen, J. Bernhardt, and R. L. Huganir, Characterization of multiple phosphorylation sites on the AMPA receptor GluR1 subunit, Neuron, vol.16, pp.1179-1188, 1996.

R. F. Rosen, B. J. Ciliax, T. S. Wingo, M. Gearing, J. Dooyema et al., Deficient High-Affinity Binding of Pittsburgh Compound B in a Case of Alzheimer's Disease, Acta neuropathological, vol.119, p.221, 2010.

W. G. Rosen, R. C. Mohs, and K. L. Davis, A new rating scale for Alzheimer's disease, Am J Psychiatry, vol.141, pp.1356-1364, 1984.

C. Rosenmund, Y. Stern-bach, and C. F. Stevens, The Tetrameric Structure of a Glutamate Receptor Channel, Science, vol.280, pp.1596-1599, 1998.

J. M. Rosenstein, J. M. Krum, and C. Ruhrberg, VEGF in the nervous system, Organogenesis, vol.6, pp.107-114, 2010.

J. M. Rosenstein, N. Mani, A. Khaibullina, and J. M. Krum, Neurotrophic Effects of Vascular Endothelial Growth Factor on Organotypic Cortical Explants and Primary Cortical Neurons, J. Neurosci, vol.23, pp.11036-11044, 2003.

D. Roth, M. Piekarek, M. Paulsson, H. Christ, W. Bloch et al., Plasmin Modulates Vascular Endothelial Growth Factor-AMediated Angiogenesis during Wound Repair, Am J Pathol, vol.168, pp.670-684, 2006.

C. Ruch, G. Skiniotis, M. O. Steinmetz, T. Walz, and K. Ballmer-hofer, Structure of a VEGF-VEGF receptor complex determined by electron microscopy, Nature Structural & Molecular Biology, vol.14, p.249, 2007.

C. Ruiz-de-almodovar, C. Coulon, P. A. Salin, E. Knevels, N. Chounlamountri et al., MatrixBinding Vascular Endothelial Growth Factor (VEGF) Isoforms Guide Granule Cell Migration in the Cerebellum via VEGF Receptor Flk1, J. Neurosci, vol.30, pp.15052-15066, 2010.

C. Ruiz-de-almodovar, D. Lambrechts, M. Mazzone, and P. Carmeliet, Role and Therapeutic Potential of VEGF in the Nervous System, Physiological Reviews, vol.89, pp.607-648, 2009.

J. Rumi-masante, F. I. Rusinga, T. E. Lester, T. B. Dunlap, T. D. Williams et al., Structural basis for activation of calcineurin by calmodulin, J. Mol. Biol, vol.415, pp.307-317, 2012.

B. K. Rycroft and A. J. Gibb, Inhibitory interactions of calcineurin (phosphatase 2B) and calmodulin on rat hippocampal NMDA receptors, Neuropharmacology, vol.47, pp.505-514, 2004.

J. K. Ryu, T. Cho, H. B. Choi, Y. T. Wang, and J. G. Mclarnon, Microglial VEGF receptor response is an integral chemotactic component in Alzheimer's disease pathology, J. Neurosci, vol.29, pp.3-13, 2009.

K. R. Sadleir, P. C. Kandalepas, V. Buggia-prévot, D. A. Nicholson, G. Thinakaran et al., Presynaptic dystrophic neurites surrounding amyloid plaques are sites of microtubule disruption, BACE1 elevation, and increased A? generation in Alzheimer's disease, Acta Neuropathol, vol.132, pp.235-256, 2016.

M. Sakono and T. Zako, Amyloid oligomers: formation and toxicity of A? oligomers, FEBS Journal, vol.277, pp.1348-1358, 2010.

Y. Sakurai, K. Ohgimoto, Y. Kataoka, N. Yoshida, and M. Shibuya, Essential role of Flk-1 (VEGF receptor 2) tyrosine residue 1173 in vasculogenesis in mice, Proc Natl Acad Sci U S A, vol.102, pp.1076-1081, 2005.

A. Salikhova, L. Wang, A. A. Lanahan, M. Liu, M. Simons et al., Vascular Endothelial Growth Factor and Semaphorin Induce Neuropilin-1 Endocytosis via Separate Pathways, Circ Res, vol.103, pp.71-79, 2008.

G. S. Salvesen, Caspases: opening the boxes and interpreting the arrows, Cell Death and Differentiation, vol.9, pp.3-5, 2002.

A. Sanz-clemente, R. A. Nicoll, and K. W. Roche, Diversity in NMDA receptor composition: many regulators, many consequences. The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry, vol.19, p.62, 2013.

S. Sarabipour, K. Ballmer-hofer, and K. Hristova, VEGFR-2 conformational switch in response to ligand binding. eLife, vol.5, p.13876, 2016.

L. P. Savtchenko and D. A. Rusakov, The optimal height of the synaptic cleft, vol.104, pp.1823-1828, 2007.

S. Sawamiphak, S. Seidel, C. L. Essmann, G. A. Wilkinson, M. E. Pitulescu et al., Ephrin-B2 regulates VEGFR2 function in developmental and tumour angiogenesis, Nature, vol.465, pp.487-491, 2010.

R. H. Scannevin and R. L. Huganir, Postsynaptic organization and regulation of excitatory synapses, Nat. Rev. Neurosci, vol.1, pp.133-141, 2000.

Q. Schwarz, C. Gu, H. Fujisawa, K. Sabelko, M. Gertsenstein et al., Vascular endothelial growth factor controls neuronal migration and cooperates with Sema3A to pattern distinct compartments of the facial nerve, Genes Dev, vol.18, pp.2822-2834, 2004.

D. J. Selkoe, Alzheimer's disease: genes, proteins, and therapy, Physiol. Rev, vol.81, pp.741-766, 2001.

D. R. Senger, S. J. Galli, A. M. Dvorak, C. A. Perruzzi, V. S. Harvey et al., Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid, Science, vol.219, pp.983-985, 1983.

A. Serrano-pozo, M. P. Frosch, E. Masliah, and B. T. Hyman, Neuropathological Alterations in Alzheimer Disease, Cold Spring Harb Perspect Med, vol.1, issue.1, p.6189, 2011.

P. Seubert, C. Vigo-pelfrey, F. Esch, M. Lee, H. Dovey et al., Isolation and quantification of soluble Alzheimer's ?-peptide from biological fluids, Nature, vol.359, pp.325-327, 1992.

F. Shalaby, J. Ho, W. L. Stanford, K. Fischer, A. C. Schuh et al., A Requirement for Flk1 in Primitive and Definitive Hematopoiesis and Vasculogenesis, Cell, vol.89, pp.981-990, 1997.

F. Shalaby, J. Rossant, T. P. Yamaguchi, M. Gertsenstein, X. F. Wu et al., Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice, Nature, vol.376, pp.62-66, 1995.

G. M. Shankar, B. L. Bloodgood, M. Townsend, D. M. Walsh, D. J. Selkoe et al., Natural Oligomers of the Alzheimer Amyloid-? 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. Shankar, S. Li, T. H. Mehta, A. Garcia-munoz, N. E. Shepardson et al., Amyloid-? protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory, Nature Medicine, vol.14, pp.837-842, 2008.

S. Shi, Y. Hayashi, R. S. Petralia, S. H. Zaman, R. J. Wenthold et al., Rapid Spine Delivery and Redistribution of AMPA Receptors After Synaptic NMDA Receptor Activation, Science, vol.284, pp.1811-1816, 1999.

M. Shibuya, Featured: Vascular endothelial growth factor and its receptor system: physiological functions in angiogenesis and pathological roles in various diseases, Journal of Biochemistry, vol.153, p.13, 2013.

M. Shibuya and L. Claessonwelsh, Signal transduction by VEGF receptors in regulation of angiogenesis and lymphangiogenesis, Experimental Cell Research, vol.312, pp.549-560, 2006.

H. Shimizu, M. Ghazizadeh, S. Sato, T. Oguro, and O. Kawanami, Interaction between ?-amyloid protein and heparan sulfate proteoglycans from the cerebral capillary basement membrane in Alzheimer's disease, Journal of Clinical Neuroscience, vol.16, pp.277-282, 2009.

A. Shinkai, M. Ito, H. Anazawa, S. Yamaguchi, K. Shitara et al., Mapping of the sites involved in ligand association and dissociation at the extracellular domain of the kinase insert domain-containing receptor for vascular endothelial growth factor, J. Biol. Chem, vol.273, pp.31283-31288, 1998.

N. Shioda, S. Moriguchi, Y. Shirasaki, and K. Fukunaga, Generation of constitutively active calcineurin by calpain contributes to delayed neuronal death following mouse brain ischemia, Journal of Neurochemistry, vol.98, pp.310-320, 2006.

T. S. Sihra, G. Flores, and A. Rodríguez-moreno, Kainate Receptors: Multiple Roles in Neuronal Plasticity, The Neuroscientist, 2013.

W. F. Silverman, J. M. Krum, N. Mani, and J. M. Rosenstein, Vascular, glial and neuronal effects of vascular endothelial growth factor in mesencephalic explant cultures, Neuroscience, vol.90, pp.1529-1541, 1999.

O. Simakova and N. J. Arispe, The cell-selective neurotoxicity of the Alzheimer's Abeta peptide is determined by surface phosphatidylserine and cytosolic ATP levels. Membrane binding is required for Abeta toxicity, J. Neurosci, vol.27, pp.13719-13729, 2007.

M. Simons, E. Gordon, and L. Claesson-welsh, Mechanisms and regulation of endothelial VEGF receptor signalling, Nature Reviews Molecular Cell Biology, vol.17, pp.611-625, 2016.

Z. ?i?ková, D. Justus, H. Kaneko, D. Friedrichs, N. Henneberg et al., Dendritic Structural Degeneration Is Functionally Linked to Cellular Hyperexcitability in a Mouse Model of Alzheimer's Disease, Neuron, vol.84, pp.1023-1033, 2014.

V. A. Skeberdis, J. Lan, T. Opitz, X. Zheng, M. V. Bennett et al., mGluR1-mediated potentiation of NMDA receptors involves a rise in intracellular calcium and activation of protein kinase C, Neuropharmacology, vol.40, pp.856-865, 2001.

F. Sladeczek, A. Momiyama, and T. Takahashi, Presynaptic Inhibitory Action of a Metabotropio Glutamate Receptor Agonist on Excitatory Transmission in Visual Cortical Neurons, Proceedings: Biological Sciences, vol.253, pp.297-303, 1993.

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

G. L. Snyder, S. Galdi, A. A. Fienberg, P. Allen, A. C. Nairn et al., Regulation of AMPA receptor dephosphorylation by glutamate receptor agonists, Neuropharmacology, vol.45, pp.703-713, 2003.

S. Soker, S. Takashima, H. Q. Miao, G. Neufeld, and M. Klagsbrun, Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor, Cell, vol.92, pp.735-745, 1998.

P. Sompol, J. L. Furman, M. M. Pleiss, S. D. Kraner, I. A. Artiushin et al., Calcineurin/NFAT Signaling in Activated Astrocytes Drives Network Hyperexcitability in A?-Bearing Mice, J. Neurosci, vol.37, pp.6132-6148, 2017.

M. Sondell, G. Lundborg, and M. Kanje, Vascular endothelial growth factor has neurotrophic activity and stimulates axonal outgrowth, enhancing cell survival and Schwann cell proliferation in the peripheral nervous system, The Journal of neuroscience, vol.19, pp.5731-5740, 1999.

Q. Song, H. Zheng, X. Li, R. L. Huganir, T. Kuner et al., Selective phosphorylation of AMPA receptor contributes to the network of long-term potentiation in the anterior cingulate cortex, The Journal of Neuroscience, pp.925-942, 2017.

Z. Songyang, S. E. Shoelson, M. Chaudhuri, G. Gish, T. Pawson et al., SH2 domains recognize specific phosphopeptide sequences, Cell, vol.72, pp.767-778, 1993.

C. Sotelo, Purkinje cell ontogeny: formation and maintenance of spines. Prog, Brain Res, vol.48, pp.149-170, 1978.

C. Sotelo, Cerebellar synaptogenesis: what we can learn from mutant mice, J. Exp. Biol, vol.153, pp.225-249, 1990.

S. F. Spampinato, S. Merlo, Y. Sano, T. Kanda, and M. A. Sortino, Astrocytes contribute to A?-induced blood-brain barrier damage through activation of endothelial MMP9, Journal of Neurochemistry, vol.142, pp.464-477, 2017.

T. L. Spires-jones and B. T. Hyman, The intersection of amyloid beta and tau at synapses in Alzheimer's disease, Neuron, vol.82, pp.756-771, 2014.

C. Spuch, D. Antequera, A. Portero, G. Orive, R. M. Hernández et al., The effect of encapsulated VEGF-secreting cells on brain amyloid load and behavioral impairment in a mouse model of Alzheimer's disease, Biomaterials, vol.31, pp.5608-5618, 2010.

S. A. Stacker, C. Caesar, M. E. Baldwin, G. E. Thornton, R. A. Williams et al., VEGF-D promotes the metastatic spread of tumor cells via the lymphatics, Nat. Med, vol.7, pp.186-191, 2001.

I. Stalmans, Y. Ng, R. Rohan, M. Fruttiger, A. Bouché et al., Arteriolar and venular patterning in retinas of mice selectively expressing VEGF isoforms, J Clin Invest, vol.109, pp.327-336, 2002.

O. Steward and P. F. Worley, A cellular mechanism for targeting newly synthesized mRNAs to synaptic sites on dendrites, vol.98, pp.7062-7068, 2001.

W. B. Stine, K. N. Dahlgren, G. A. Krafft, and M. J. Ladu, In Vitro Characterization of Conditions for Amyloid-? Peptide Oligomerization and Fibrillogenesis, J. Biol. Chem, vol.278, pp.11612-11622, 2003.

J. Stöhr, C. Condello, J. C. Watts, L. Bloch, A. Oehler et al., Distinct synthetic A? prion strains producing different amyloid deposits in bigenic mice, vol.111, pp.10329-10334, 2014.

J. Stohr, J. C. Watts, Z. L. Mensinger, A. Oehler, S. K. Grillo et al., Purified and synthetic Alzheimer's amyloid beta (A ) prions, PNAS, vol.109, pp.11025-11030, 2012.

X. Sun, Y. Wu, B. Chen, Z. Zhang, W. Zhou et al., Regulator of calcineurin 1 (RCAN1) facilitates neuronal apoptosis through caspase 3 activation, J. Biol. Chem, vol.286, pp.9049-9062, 2011.

Z. Sun, X. Li, S. Massena, S. Kutschera, N. Padhan et al., VEGFR2 induces c-Src signaling and vascular permeability in vivo via the adaptor protein TSAd, J Exp Med, vol.209, pp.1363-1377, 2012.

N. Suzuki, T. T. Cheung, X. D. Cai, A. Odaka, L. Otvos et al., An increased percentage of long amyloid beta protein secreted by familial amyloid beta protein precursor (beta APP717) mutants, Science, vol.264, pp.1336-1340, 1994.

T. Suzuki and K. Okumura-noji, NMDA receptor subunits epsilon 1 (NR2A) and epsilon 2 (NR2B) are substrates for Fyn in the postsynaptic density fraction isolated from the rat brain, Biochem. Biophys. Res. Commun, vol.216, pp.582-588, 1995.

N. Tae, S. Lee, O. Kim, J. Park, S. Na et al., Syntenin promotes VEGFinduced VEGFR2 endocytosis and angiogenesis by increasing ephrin-B2 function in endothelial cells, Oncotarget, vol.8, pp.38886-38901, 2017.

G. Taglialatela, D. Hogan, W. Zhang, and K. T. Dineley, Intermediate-and LongTerm Recognition Memory Deficits in Tg2576 Mice Are Reversed with Acute Calcineurin Inhibition, Behav. Brain Res, vol.200, pp.95-99, 2009.

T. Takadera and T. Ohyashiki, Caspase-dependent apoptosis induced by calcineurin inhibitors was prevented by glycogen synthase kinase-3 inhibitors in cultured rat cortical cells, Brain Research, vol.1133, pp.20-26, 2007.

T. Takahashi and M. Shibuya, The 230 kDa mature form of KDR/Flk-1 (VEGF receptor-2) activates the PLC-gamma pathway and partially induces mitotic signals in NIH3T3 fibroblasts, Oncogene, vol.14, pp.2079-2089, 1997.

T. Takahashi, H. Ueno, and M. Shibuya, VEGF activates protein kinase C-dependent, but Ras-independent Raf-MEK-MAP kinase pathway for DNA synthesis in primary endothelial cells, Oncogene, vol.18, pp.2221-2230, 1999.

Y. Takumi, V. Ramírez-león, P. Laake, E. Rinvik, and O. P. Ottersen, Different modes of expression of AMPA and NMDA receptors in hippocampal synapses, Nature Neuroscience, vol.2, p.618, 1999.

M. Talantova, S. Sanz-blasco, X. Zhang, P. Xia, M. W. Akhtar et al., A? induces astrocytic glutamate release, extrasynaptic NMDA receptor activation, and synaptic loss, vol.110, pp.2518-2527, 2013.

J. Tanaka, Y. Horiike, M. Matsuzaki, T. Miyazaki, G. C. Ellis-davies et al., Protein Synthesis and Neurotrophin-Dependent Structural Plasticity of Single Dendritic Spines, Science, vol.319, pp.1683-1687, 2008.

H. Tang, X. Mao, L. Xie, D. A. Greenberg, and K. Jin, Expression Level of Vascular Endothelial Growth Factor in Hippocampus is Associated with Cognitive Impairment in Patients with Alzheimer's Disease, Neurobiol Aging, vol.34, pp.1412-1415, 2013.

R. K. Tannenberg, H. L. Scott, A. E. Tannenberg, and P. R. Dodd, Selective loss of synaptic proteins in Alzheimer's disease: evidence for an increased severity with APOE varepsilon4, Neurochem. Int, vol.49, pp.631-639, 2006.

,

E. Tarkowski, R. Issa, M. Sjogren, A. Wallin, K. Blennow et al., Increased intrathecal levels of the angiogenic factors VEGF and TGF-in Alzheimer's disease and vascular dementia, Neurobiology of Aging, p.7, 2002.

A. Tashiro, A. Minden, and R. Yuste, Regulation of dendritic spine morphology by the rho family of small GTPases: antagonistic roles of Rac and Rho, Cereb. Cortex, vol.10, pp.927-938, 2000.

R. D. Terry, E. Masliah, D. P. Salmon, N. Butters, R. Deteresa et al., Physical basis of cognitive alterations in alzheimer's disease: Synapse loss is the major correlate of cognitive impairment, Ann Neurol, vol.30, pp.572-580, 1991.

G. Tesco, Y. H. Koh, E. Kang, A. Cameron, S. Das et al., Depletion of GGA3 stabilizes BACE and enhances ?-secretase activity, Neuron, vol.54, pp.721-737, 2007.

L. Texidó, M. Martín-satué, E. Alberdi, C. Solsona, and C. Matute, Amyloid ? peptide oligomers directly activate NMDA receptors, Cell Calcium, vol.49, pp.184-190, 2011.

D. R. Thal, U. Rüb, M. Orantes, and H. Braak, Phases of Abeta-deposition in the human brain and its relevance for the development of AD, Neurology, vol.58, issue.12, pp.1791-800, 2002.

T. Thomas, S. Miners, and S. Love, Post-mortem assessment of hypoperfusion of cerebral cortex in Alzheimer's disease and vascular dementia, Brain, vol.138, pp.1059-1069, 2015.

W. F. Timmers, F. Tagliavini, J. Haan, and B. Frangione, Parenchymal preamyloid and amyloid deposits in the brains of patients with hereditary cerebral hemorrhage with amyloidosis--Dutch type, Neurosci. Lett, vol.118, pp.223-226, 1990.

T. Tobimatsu and H. Fujisawa, Tissue-specific expression of four types of rat calmodulin-dependent protein kinase II mRNAs, J. Biol. Chem, vol.264, pp.17907-17912, 1989.

S. E. Tom, R. A. Hubbard, P. K. Crane, S. J. Haneuse, J. Bowen et al., Characterization of Dementia and Alzheimer's Disease in an Older Population: Updated Incidence and Life Expectancy With and Without Dementia, Am J Public Health, vol.105, pp.408-413, 2014.

G. Tong, D. Shepherd, J. , and C. , Synaptic desensitization of NMDA receptors by calcineurin, Science, vol.267, pp.1510-1512, 1995.

M. Townsend, G. M. Shankar, T. Mehta, D. M. Walsh, and D. J. Selkoe, Effects of secreted oligomers of amyloid ?-protein on hippocampal synaptic plasticity: a potent role for trimers: Amyloid ?-protein and hippocampal synaptic plasticity, The Journal of Physiology, vol.572, pp.477-492, 2006.

S. F. Traynelis, L. P. Wollmuth, C. J. Mcbain, F. S. Menniti, K. M. Vance et al., Glutamate receptor ion channels: structure, regulation, and function, Pharmacol. Rev, vol.62, pp.405-496, 2010.

J. W. Um, H. B. Nygaard, J. K. Heiss, M. A. Kostylev, M. Stagi et al., Alzheimer amyloid-? oligomer bound to postsynaptic prion protein activates Fyn to impair neurons, Nat Neurosci, vol.15, pp.1227-1235, 2012.

B. Urbanc, L. Cruz, R. Le, J. Sanders, K. H. Ashe et al., Neurotoxic effects of thioflavin S-positive amyloid deposits in transgenic mice and Alzheimer's disease, PNAS, vol.99, pp.13990-13995, 2002.

R. Usui, M. Shibuya, S. Ishibashi, and Y. Maru, Ligand-independent activation of vascular endothelial growth factor receptor 1 by low-density lipoprotein, EMBO Rep, vol.8, pp.1155-1161, 2007.

J. E. Vaughn, Fine structure of synaptogenesis in the vertebrate central nervous system, Synapse, vol.3, pp.255-285, 1989.

A. K. Vehmas, C. H. Kawas, W. F. Stewart, and J. C. Troncoso, Immune reactive cells in senile plaques and cognitive decline in Alzheimer's disease, Neurobiol. of Aging, vol.24, pp.321-331, 2003.

B. Vellas, N. Coley, P. Ousset, G. Berrut, J. Dartigues et al., Long-term use of standardised Ginkgo biloba extract for the prevention of Alzheimer's disease (GuidAge): a randomised placebo-controlled trial, Lancet Neurol, vol.11, pp.851-859, 2012.

A. Volianskis, G. L. Collingridge, and M. S. Jensen, The roles of STP and LTP in synaptic encoding, Peerj, vol.1, p.3, 2013.

V. Rotz and R. C. , The APP intracellular domain forms nuclear multiprotein complexes and regulates the transcription of its own precursor, Journal of Cell Science, vol.117, pp.4435-4448, 2004.

D. M. Walsh, I. Klyubin, J. V. Fadeeva, W. K. Cullen, R. Anwyl et al., Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo, Nature, vol.416, pp.535-539, 2002.

G. Wang, J. Gilbert, and H. Man, AMPA Receptor Trafficking in Homeostatic Synaptic Plasticity: Functional Molecules and Signaling Cascades, Neural. Plast, p.825364, 2012.

Y. Wang, G. Chen, X. Hu, Y. Lu, J. Zhou et al., The expression of calcium/calmodulin-dependent protein kinase II-alpha in the hippocampus of patients with Alzheimer's disease and its links with AD-related pathology, Brain Res, vol.1031, pp.101-108, 2005.

S. J. Webster, A. D. Bachstetter, P. T. Nelson, F. A. Schmitt, and L. J. Van-eldik, Using mice to model Alzheimer's dementia: an overview of the clinical disease and the preclinical behavioral changes in 10 mouse models, p.5, 2014.

J. C. Weddell, S. Chen, and P. I. Imoukhuede, VEGFR1 promotes cell migration and proliferation through PLC? and PI3K pathways, Systems Biology and Applications, vol.4, p.1, 2017.

M. Weider, L. J. Starost, K. Groll, M. Küspert, E. Sock et al., Nfat/calcineurin signaling promotes oligodendrocyte differentiation and myelination by transcription factor network tuning, Nat. Commun, vol.9, p.899, 2018.

S. Weis, J. Cui, L. Barnes, and D. Cheresh, Endothelial barrier disruption by VEGFmediated Src activity potentiates tumor cell extravasation and metastasis, J Cell Biol, vol.167, pp.223-229, 2004.

W. Weng, K. Lin, P. Wu, Y. Ho, Y. Liu et al., VEGF expression correlates with neuronal differentiation and predicts a favorable prognosis in patients with neuroblastoma, Scientific Reports, vol.7, p.11212, 2017.

M. Q. Wentink, T. M. Hackeng, S. P. Tabruyn, W. C. Puijk, K. Schwamborn et al., Targeted vaccination against the bevacizumab binding site on VEGF using 3D-structured peptides elicits efficient antitumor activity, vol.113, pp.12532-12537, 2016.

X. Q. Werdich and J. S. Penn, Src, Fyn and Yes play differential roles in VEGF-mediated endothelial cell events, Angiogenesis, vol.8, pp.315-326, 2005.

K. C. Wheeler, M. K. Jena, B. S. Pradhan, N. Nayak, S. Das et al., VEGF may contribute to macrophage recruitment and M2 polarization in the decidua, PLoS One, vol.13, 2018.

J. R. Wild, C. A. Staton, K. Chapple, and B. M. Corfe, Neuropilins: expression and roles in the epithelium, Int J Exp Pathol, vol.93, pp.81-103, 2012.

M. Willem, S. Tahirovic, M. A. Busche, S. V. Ovsepian, M. Chafai et al., Nature, vol.526, pp.443-447, 2015.

T. Wisniewski, A. Golabek, T. Vogel, and B. Frangione, Acceleration of Alzheimer's Fibril Formation by Apolipoprotein E In Vitro, Am J Pathol, vol.145, p.6, 1994.

I. M. Wittko, A. Schanzer, A. Kuzmichev, F. T. Schneider, M. Shibuya et al., VEGFR-1 Regulates Adult Olfactory Bulb Neurogenesis and Migration of Neural Progenitors in the Rostral Migratory Stream In Vivo, Journal of Neuroscience, vol.29, pp.8704-8714, 2009.

J. Woolard, W. Wang, H. S. Bevan, Y. Qiu, L. Morbidelli et al., VEGF165b, an inhibitory vascular endothelial growth factor splice variant: mechanism of action, Cancer Res, vol.64, pp.7822-7835, 2004.

S. Wray and W. Noble, Linking Amyloid and Tau Pathology in Alzheimer's Disease: The Role of Membrane Cholesterol in A?-Mediated Tau Toxicity, J. Neurosci, vol.29, p.9665, 2009.

H. Wu, E. Hudry, T. Hashimoto, K. Kuchibhotla, A. Rozkalne et al., , 2010.

, A?) Induces the Morphological Neurodegenerative Triad of Spine Loss, Dendritic Simplification, and Neuritic Dystrophies through Calcineurin (CaN) Activation, Amyloid Beta, vol.30, pp.2636-2649

H. Wu, K. Tomizawa, Y. Oda, F. Wei, Y. Lu et al., Critical Role of Calpain-mediated Cleavage of Calcineurin in Excitotoxic Neurodegeneration, J. Biol. Chem, vol.279, pp.4929-4940, 2004.

P. Xia, L. P. Aiello, H. Ishii, Z. Y. Jiang, D. J. Park et al., Characterization of vascular endothelial growth factor's effect on the activation of protein kinase C, its isoforms, and endothelial cell growth, J Clin Invest, vol.98, pp.2018-2026, 1996.

Z. Xie, D. P. Srivastava, H. Photowala, L. Kai, M. E. Cahill et al., Kalirin-7 Controls Activity-Dependent Structural and Functional Plasticity of Dendritic Spines, Neuron, vol.56, pp.640-656, 2007.

S. D. Yan, A. Roher, A. M. Schmidt, and D. M. Stern, Cellular cofactors for amyloid betapeptide-induced cell stress. Moving from cell culture to in vivo, Am. J. Pathol, vol.155, pp.1403-1411, 1999.

Y. Yan and C. Wang, A?42 is More Rigid than A?40 at the C Terminus: Implications for A? Aggregation and Toxicity, Journal of Molecular Biology, vol.364, pp.853-862, 2006.

S. Yang, D. Bae, H. J. Kang, B. J. Gwag, Y. S. Gho et al., Coaccumulation of vascular endothelial growth factor with ?-amyloid in the brain of patients with Alzheimer's disease, Neurobiology of Aging, vol.25, pp.283-290, 2004.

S. Yang, L. Zhang, Y. Huang, and F. Sun, Distribution of Flk-1 and Flt-1 receptors in neonatal and adult rat brains, The Anatomical Record Part A: Discoveries in Molecular, Cellular, and Evolutionary Biology, vol.274, pp.851-856, 2003.

Y. Yao, J. Belcher, A. J. Berger, M. L. Mayer, and A. Y. Lau, Conformational Analysis of NMDA Receptor GluN1, GluN2, and GluN3 Ligand-Binding Domains Reveals Subtype-Specific Characteristics. Structure, vol.8, 2013.

Y. Yao and E. J. Duh, VEGF selectively induces Down syndrome critical region 1 gene expression in endothelial cells: a mechanism for feedback regulation of angiogenesis?, Biochem. Biophys. Res. Commu, vol.321, pp.648-656, 2004.

N. Yasumatsu, M. Matsuzaki, T. Miyazaki, J. Noguchi, and H. Kasai, Principles of Long-Term Dynamics of Dendritic Spines, J. Neurosci, vol.28, pp.13592-13608, 2008.

Y. Yin, D. Gao, Y. Wang, Z. Wang, X. Wang et al., Tau accumulation induces synaptic impairment and memory deficit by calcineurin-mediated inactivation of nuclear CaMKIV/CREB signaling, vol.113, pp.3773-3781, 2016.

S. G. Younkin, Evidence that A beta 42 is the real culprit in Alzheimer's disease, Ann. Neurol, vol.37, pp.287-288, 1995.

L. Yuan, D. Moyon, L. Pardanaud, C. Bréant, M. J. Karkkainen et al., Abnormal lymphatic vessel development in neuropilin 2 mutant mice, Development, vol.129, pp.4797-4806, 2002.

R. Yuste and T. Bonhoeffer, Genesis of dendritic spines: insights from ultrastructural and imaging studies, Nature Reviews Neuroscience, vol.5, pp.24-34, 2004.

R. Yuste, A. Majewska, S. S. Cash, and W. Denk, Mechanisms of Calcium Influx into Hippocampal Spines: Heterogeneity among Spines, Coincidence Detection by NMDA Receptors, and Optical Quantal Analysis, J. Neurosci, vol.19, pp.1976-1987, 1999.

I. Zachary and G. Gliki, Signaling transduction mechanisms mediating biological actions of the vascular endothelial growth factor family, Cardiovasc. Res, vol.49, pp.568-581, 2001.

S. Zechel, K. Unsicker, and O. Von-bohlen-und-halbach, Fibroblast growth factor-2 deficiency affects hippocampal spine morphology, but not hippocampal catecholaminergic or cholinergic innervation, Developmental Dynamics, vol.238, pp.343-350, 2009.

Y. Zeng, D. Zhao, and C. Xie, Neurotrophins Enhance CaMKII Activity and Rescue Amyloid-?-Induced Deficits in Hippocampal Synaptic Plasticity, Journal of Alzheimer's Disease, vol.21, pp.823-831, 2010.

L. Zhang, Y. Fang, Y. Xu, Y. Lian, N. Xie et al., Curcumin Improves Amyloid ?-Peptide (1-42) Induced Spatial Memory Deficits through BDNF-ERK Signaling Pathway, PLOS ONE, vol.10, p.131525, 2015.

L. Zhang, F. Zhou, W. Han, B. Shen, J. Luo et al., VEGFR-3 ligand-binding and kinase activity are required for lymphangiogenesis but not for angiogenesis, Cell Res, vol.20, pp.1319-1331, 2010.

W. Zhang, J. Hao, R. Liu, Z. Zhang, G. Lei et al., Soluble A? levels correlate with cognitive deficits in the 12-month-old APPswe/PS1dE9 mouse model of Alzheimer's disease, Behav. Brain Res, vol.222, pp.342-350, 2011.

W. Zhang, P. J. Ross, Y. Tu, Y. Wang, S. Beggs et al., Fyn Kinase regulates GluN2B subunit-dominant NMDA receptors in human induced pluripotent stem cell-derived neurons, Scientific Reports, vol.6, p.23837, 2016.

X. Zhang and W. Song, The role of APP and BACE1 trafficking in APP processing and amyloid-? generation, Alzheimers Res Ther, vol.5, p.46, 2013.

Y. Zhang, N. Holbro, and T. G. Oertner, Optical induction of plasticity at single synapses reveals input-specific accumulation of ?CaMKII, vol.105, pp.12039-12044, 2008.

Z. Zheng, B. Sabirzhanov, and J. Keifer, Oligomeric amyloid-{beta} inhibits the proteolytic conversion of brain-derived neurotrophic factor (BDNF), AMPA receptor trafficking, and classical conditioning, J. Biol. Chem, vol.285, pp.34708-34717, 2010.

Q. Zhou, K. J. Homma, and M. Poo, Shrinkage of dendritic spines associated with longterm depression of hippocampal synapses, Neuron, vol.44, pp.749-757, 2004.

N. E. Ziv and S. J. Smith, Evidence for a role of dendritic filopodia in synaptogenesis and spine formation, Neuron, vol.17, pp.91-102, 1996.