B. S. Abrahams, D. E. Arking, D. B. Campbell, H. C. Mefford, E. M. Morrow et al., SFARI Gene 2.0: a community-driven knowledgebase for the autism spectrum disorders (ASDs), Molecular autism, vol.4, issue.1, p.3851189, 2013.

E. J. Lee, S. Y. Choi, and E. Kim, NMDA receptor dysfunction in autism spectrum disorders, Curr Opin Pharmacol, vol.20, pp.8-13, 2015.

K. B. Hansen, F. Yi, R. E. Perszyk, H. Furukawa, L. P. Wollmuth et al., Structure, function, and allosteric modulation of NMDA receptors, The Journal of general physiology, vol.150, issue.8, p.6080888, 2018.

S. Zhu and P. Paoletti, Allosteric modulators of NMDA receptors: multiple sites and mechanisms. Current opinion in pharmacology, vol.20, pp.14-23, 2015.

P. Paoletti, C. Bellone, and Q. Zhou, NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease, Nature reviews Neuroscience, vol.14, issue.6, pp.383-400, 2013.

M. P. Lussier, A. Sanz-clemente, and K. W. Roche, Dynamic Regulation of N-Methyl-d-aspartate (NMDA) and alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA) Receptors by Posttranslational Modifications, The Journal of biological chemistry, vol.290, issue.48, pp.28596-603, 2015.

C. G. Lau and R. S. Zukin, NMDA receptor trafficking in synaptic plasticity and neuropsychiatric disorders, Nature reviews Neuroscience, vol.8, issue.6, pp.413-439, 2007.

W. Chung, S. Y. Choi, E. Lee, H. Park, J. Kang et al., Social deficits in IRSp53 mutant mice improved by NMDAR and mGluR5 suppression. Nature neuroscience, 2015.

H. Won, H. R. Lee, H. Y. Gee, W. Mah, J. I. Kim et al., Autistic-like social behaviour in Shank2-mutant mice improved by restoring NMDA receptor function, Nature, vol.486, issue.7402, pp.261-266, 2012.

J. Blundell, C. A. Blaiss, M. R. Etherton, F. Espinosa, K. Tabuchi et al., Neuroligin-1 deletion results in impaired spatial memory and increased repetitive behavior, The Journal of neuroscience, vol.30, issue.6, 2010.

R. A. Myers, F. Casals, J. Gauthier, F. F. Hamdan, J. Keebler et al., A population genetic approach to mapping neurological disorder genes using deep resequencing, PLoS Genet, vol.7, issue.2, 2011.

B. J. O'roak, L. Vives, W. Fu, J. D. Egertson, I. B. Stanaway et al., Multiplex targeted sequencing identifies recurrently mutated genes in autism spectrum disorders, Science, vol.338, issue.6114, p.3528801, 2012.

S. De-rubeis, X. He, A. P. Goldberg, C. S. Poultney, K. Samocha et al., Synaptic, transcriptional and chromatin genes disrupted in autism, Nature, vol.515, issue.7526, p.4402723, 2014.

I. Iossifov, D. Levy, J. Allen, K. Ye, M. Ronemus et al., Low load for disruptive mutations in autism genes and their biased transmission, Proceedings of the National Academy of Sciences of the United States of America, vol.112, issue.41, pp.5600-5607, 2015.

K. Platzer, H. Yuan, H. Schutz, A. Winschel, W. Chen et al., GRIN2B encephalopathy: novel findings on phenotype, variant clustering, functional consequences and treatment aspects, Journal of medical genetics, vol.54, issue.7, pp.460-70, 2017.

P. Central and P. , , p.5656050

H. J. Yoo, I. H. Cho, M. Park, S. Y. Yang, and S. A. Kim, Family based association of GRIN2A and GRIN2B with Korean autism spectrum disorders, Neurosci Lett, vol.512, issue.2, pp.89-93, 2012.

C. Hu, W. Chen, S. J. Myers, H. Yuan, and S. F. Traynelis, Human GRIN2B variants in neurodevelopmental disorders, Journal of pharmacological sciences, vol.132, issue.2, p.5125235, 2016.

Y. P. Tang, E. Shimizu, G. R. Dube, C. Rampon, G. A. Kerchner et al., Genetic enhancement of learning and memory in mice, Nature, vol.401, issue.6748, pp.63-72, 1999.

W. Xiangwei, Y. Jiang, and H. Yuan, De Novo Mutations and Rare Variants Occurring in NMDA Receptors, Curr Opin Physiol, vol.2, pp.27-35, 2018.

P. Central and P. , , p.5945193

S. A. Swanger, W. Chen, G. Wells, P. B. Burger, A. Tankovic et al., Mechanistic Insight into NMDA Receptor Dysregulation by Rare Variants in the GluN2A and GluN2B Agonist Binding Domains

, Am J Hum Genet, vol.99, issue.6, pp.1261-80, 2016.

, PubMed Central

H. Furukawa and E. Gouaux, Mechanisms of activation, inhibition and specificity: crystal structures of the NMDA receptor NR1 ligand-binding core, The EMBO journal, vol.22, issue.12, p.162155, 2003.

C. H. Lee, W. Lu, J. C. Michel, A. Goehring, J. Du et al., NMDA receptor structures reveal subunit arrangement and pore architecture, Nature, vol.511, issue.7508, p.4263351, 2014.

E. Karakas and H. Furukawa, Crystal structure of a heterotetrameric NMDA receptor ion channel, Science, vol.344, issue.6187, p.4113085, 2014.

B. Serraz, T. Grand, and P. Paoletti, Altered zinc sensitivity of NMDA receptors harboring clinically-relevant mutations, Neuropharmacology, vol.109, pp.196-204, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01331195

L. Mony, S. Zhu, S. Carvalho, and P. Paoletti, Molecular basis of positive allosteric modulation of GluN2B NMDA receptors by polyamines, The EMBO journal, vol.30, issue.15, pp.3134-3180, 2011.

J. Rachline, F. Perin-dureau, L. Goff, A. Neyton, J. Paoletti et al., The micromolar zinc-binding domain on the NMDA receptor subunit NR2B, The Journal of neuroscience, vol.25, issue.2, pp.308-325, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00115448

M. Sheng, J. Cummings, L. A. Roldan, Y. N. Jan, and J. Ly, Changing subunit composition of heteromeric NMDA receptors during development of rat cortex, Nature, vol.368, issue.6467, pp.144-151, 1994.

D. Stroebel, M. Casado, and P. Paoletti, Triheteromeric NMDA receptors: from structure to synaptic physiology, Curr Opin Physiol, vol.2, pp.1-12, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02438993

P. Central and P. , , p.5905667

T. Tada and M. Sheng, Molecular mechanisms of dendritic spine morphogenesis. Current opinion in neurobiology, vol.16, pp.95-101, 2006.

M. B. Kennedy, The postsynaptic density. Current opinion in neurobiology, vol.3, pp.732-739, 1993.

M. Sheng and C. Sala, PDZ domains and the organization of supramolecular complexes, Annual review of neuroscience, vol.24, pp.1-29, 2001.

J. A. Gray, Y. Shi, H. Usui, M. J. During, K. Sakimura et al., Distinct modes of AMPA receptor suppression at developing synapses by GluN2A and GluN2B: single-cell NMDA receptor subunit deletion in vivo, Neuron, vol.71, issue.6, pp.1085-101, 2011.

P. Central and P. , , p.3183990

G. Bouvier, C. Bidoret, M. Casado, and P. Paoletti, Presynaptic NMDA receptors: Roles and rules. Neuroscience, vol.311, pp.322-362, 2015.

F. Yi, T. Danko, S. C. Botelho, C. Patzke, C. Pak et al., Autism-associated SHANK3 haploinsufficiency causes Ih channelopathy in human neurons, Science, vol.352, issue.6286, p.4901875, 2016.

J. L. Silverman, M. Yang, C. Lord, and J. N. Crawley, Behavioural phenotyping assays for mouse models of autism, Nature reviews Neuroscience, vol.11, issue.7, p.3087436, 2010.

M. L. Scattoni, J. Crawley, and L. Ricceri, Ultrasonic vocalizations: a tool for behavioural phenotyping of mouse models of neurodevelopmental disorders, Neuroscience and biobehavioral reviews, vol.33, issue.4, p.2688771, 2009.

M. Willadsen, D. Seffer, R. K. Schwarting, and M. Wohr, Rodent ultrasonic communication: Male prosocial 50-kHz ultrasonic vocalizations elicit social approach behavior in female rats (Rattus norvegicus), Journal of comparative psychology, vol.128, issue.1, pp.56-64, 2014.

J. T. Winslow and T. R. Insel, The infant rat separation paradigm: a novel test for novel anxiolytics, Trends Pharmacol Sci, vol.12, issue.11, pp.402-406, 1991.

S. W. White, D. Oswald, T. Ollendick, and L. Scahill, Anxiety in children and adolescents with autism spectrum disorders, Clin Psychol Rev, vol.29, issue.3, p.2692135, 2009.

S. Schade, W. Paulus, and . D-cycloserine, The international journal of neuropsychopharmacology / official scientific journal of the Collegium Internationale Neuropsychopharmacologicum, Neuropsychiatric Diseases: A Systematic Review, vol.19, 2016.

P. Central and P. , , p.4851259

E. J. Lee, H. Lee, T. N. Huang, C. Chung, W. Shin et al., Trans-synaptic zinc mobilization improves social interaction in two mouse models of autism through NMDAR activation, Nat Commun, vol.6, p.4479043, 2015.

T. N. Huang, H. C. Chuang, W. H. Chou, C. Y. Chen, H. F. Wang et al., Tbr1 haploinsufficiency impairs amygdalar axonal projections and results in cognitive abnormality, Nat Neurosci, vol.17, issue.2, pp.240-247, 2014.

T. N. Huang, T. L. Yen, L. R. Qiu, H. C. Chuang, J. P. Lerch et al., Haploinsufficiency of autism causative gene Tbr1 impairs olfactory discrimination and neuronal activation of the olfactory system in mice, Molecular autism, vol.10, p.6371489, 2019.

A. M. Mabb and M. D. Ehlers, Ubiquitination in postsynaptic function and plasticity. Annual review of cell and developmental biology, vol.26, pp.179-210, 2010.

H. C. Tai and E. M. Schuman, Ubiquitin, the proteasome and protein degradation in neuronal function and dysfunction, Nature reviews Neuroscience, vol.9, issue.11, pp.826-864, 2008.

H. Kawabe and N. Brose, The role of ubiquitylation in nerve cell development, Nature reviews Neuroscience, vol.12, issue.5, pp.251-68, 2011.

J. J. Yi and M. D. Ehlers, Emerging roles for ubiquitin and protein degradation in neuronal function. Pharmacological reviews, vol.59, pp.14-39, 2007.

D. B. Scott, I. Michailidis, Y. Mu, D. Logothetis, and M. D. Ehlers, Endocytosis and degradative sorting of NMDA receptors by conserved membrane-proximal signals, The Journal of neuroscience, vol.24, issue.32, pp.7096-109, 2004.

M. J. Kennedy and M. D. Ehlers, Organelles and trafficking machinery for postsynaptic plasticity. Annual review of neuroscience, vol.29, pp.325-62, 2006.

P. L. Chazot and F. A. Stephenson, Molecular dissection of native mammalian forebrain NMDA receptors containing the NR1 C2 exon: direct demonstration of NMDA receptors comprising NR1, NR2A, and NR2B subunits within the same complex, Journal of neurochemistry, vol.69, issue.5, p.9349560, 1997.

J. Luo, Y. Wang, R. P. Yasuda, A. W. Dunah, and B. B. Wolfe, The majority of N-methyl-D-aspartate receptor complexes in adult rat cerebral cortex contain at least three different subunits (NR1/NR2A/NR2B), Molecular pharmacology, vol.51, issue.1, pp.79-86, 1997.

C. Rauner and G. Kohr, Triheteromeric NR1/NR2A/NR2B receptors constitute the major N-methyl-D-aspartate receptor population in adult hippocampal synapses, The Journal of biological chemistry, vol.286, issue.9, p.3045010, 2011.

K. R. Tovar, M. J. Mcginley, and G. L. Westbrook, Triheteromeric NMDA receptors at hippocampal synapses. The Journal of neuroscience, vol.33, pp.9150-60, 2013.

C. J. Hatton and P. Paoletti, Modulation of triheteromeric NMDA receptors by N-terminal domain ligands, Neuron, vol.46, issue.2, pp.261-74, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00139989

K. B. Hansen, K. K. Ogden, H. Yuan, and S. F. Traynelis, Distinct functional and pharmacological properties of Triheteromeric GluN1/GluN2A/GluN2B NMDA receptors, Neuron, vol.81, issue.5, p.3957490, 2014.

D. Stroebel, S. Carvalho, T. Grand, S. Zhu, and P. Paoletti, Controlling NMDA receptor subunit composition using ectopic retention signals. The Journal of neuroscience: the official journal of the Society for Neuroscience, vol.34, pp.16630-16636, 2014.

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, issue.5673, pp.1021-1025, 2004.

P. V. Massey, B. E. Johnson, P. R. Moult, Y. P. Auberson, M. W. Brown et al., Differential roles of NR2A and NR2B-containing NMDA receptors in cortical long-term potentiation and long-term depression, The Journal of neuroscience, vol.24, issue.36, pp.7821-7829, 2004.

C. J. Fox, K. I. Russell, Y. T. Wang, and B. R. Christie, Contribution of NR2A and NR2B NMDA subunits to bidirectional synaptic plasticity in the hippocampus in vivo, Hippocampus, vol.16, issue.11, pp.907-922, 2006.

Y. Izumi, Y. P. Auberson, and C. F. Zorumski, Zinc modulates bidirectional hippocampal plasticity by effects on NMDA receptors, The Journal of neuroscience, vol.26, issue.27, pp.7181-7189, 2006.

T. E. Bartlett, N. J. Bannister, V. J. Collett, S. L. Dargan, P. V. Massey et al., Differential roles of NR2A and NR2B-containing NMDA receptors in LTP and LTD in the CA1 region of two-week old rat hippocampus, Neuropharmacology, vol.52, issue.1, pp.60-70, 2007.

W. Morishita, W. Lu, G. B. Smith, R. A. Nicoll, M. F. Bear et al., Activation of NR2B-containing NMDA receptors is not required for NMDA receptor-dependent long-term depression, Neuropharmacology, vol.52, issue.1, pp.71-77, 2007.

O. A. Shipton and O. Paulsen, GluN2A and GluN2B subunit-containing NMDA receptors in hippocampal plasticity, Philosophical transactions of the Royal Society of London Series B, Biological sciences, vol.369, p.3843894, 1633.

J. Neyton and P. Paoletti, Relating NMDA receptor function to receptor subunit composition: limitations of the pharmacological approach, The Journal of neuroscience, vol.26, issue.5, pp.1331-1334, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00115285

K. R. Tovar and G. L. Westbrook, Amino-terminal ligands prolong NMDA Receptor-mediated EPSCs, The Journal of neuroscience, vol.32, issue.23, pp.8065-73, 2012.

J. N. Kew, G. Trube, and J. A. Kemp, A novel mechanism of activity-dependent NMDA receptor antagonism describes the effect of ifenprodil in rat cultured cortical neurones, The Journal of physiology, vol.497, p.1160972, 1996.

K. Dore, J. Aow, and R. Malinow, Agonist binding to the NMDA receptor drives movement of its cytoplasmic domain without ion flow, Proceedings of the National Academy of Sciences of the United States of America, vol.112, issue.47, pp.14705-14715, 2015.

P. Central and P. , , p.4664357

S. Nabavi, H. W. Kessels, S. Alfonso, J. Aow, R. Fox et al., Metabotropic NMDA receptor function is required for NMDA receptor-dependent long-term depression, Proceedings of the National Academy of Sciences of the United States of America, vol.110, issue.10, p.3593861, 2013.

I. S. Stein, J. A. Gray, and K. Zito, Non-Ionotropic NMDA Receptor Signaling Drives Activity-Induced Dendritic Spine Shrinkage, The Journal of neuroscience, vol.35, issue.35, p.4556794, 2015.

J. M. Wong and J. A. Gray, Long-Term Depression Is Independent of GluN2 Subunit Composition, The Journal of neuroscience: the official journal of the Society for Neuroscience, vol.38, p.5943974, 2018.

A. Gillott, F. Furniss, and A. Walter, Anxiety in high-functioning children with autism, Autism, vol.5, issue.3, pp.277-86, 2001.

E. Ey, C. S. Leblond, and T. Bourgeron, Behavioral profiles of mouse models for autism spectrum disorders, Autism research, vol.4, issue.1, pp.5-16, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-01470286

Y. H. Jiang and M. D. Ehlers, Modeling autism by SHANK gene mutations in mice, Neuron, vol.78, issue.1, p.3659167, 2013.

J. T. Winslow, E. F. Hearn, J. Ferguson, L. J. Young, M. M. Matzuk et al., Infant vocalization, adult aggression, and fear behavior of an oxytocin null mutant mouse, Hormones and behavior, vol.37, issue.2, pp.145-55, 2000.

A. J. Guastella and I. B. Hickie, Oxytocin Treatment, Circuitry, and Autism: A Critical Review of the Literature Placing Oxytocin Into the Autism Context. Biological psychiatry, vol.79, pp.234-276, 2016.

R. C. Malenka and M. F. Bear, LTP and LTD: an embarrassment of riches, Neuron, vol.44, issue.1, pp.5-21, 2004.

G. L. Collingridge, S. Peineau, J. G. Howland, and Y. T. Wang, Long-term depression in the CNS, Nature reviews Neuroscience, vol.11, issue.7, pp.459-73, 2010.

L. M. Ritter, D. M. Vazquez, and J. H. Meador-woodruff, Ontogeny of ionotropic glutamate receptor subunit expression in the rat hippocampus, Brain research Developmental brain research, vol.139, issue.2, pp.227-263, 2002.

N. Kemp, J. Mcqueen, S. Faulkes, and Z. I. Bashir, Different forms of LTD in the CA1 region of the hippocampus: role of age and stimulus protocol. The European journal of neuroscience, vol.12, pp.360-366, 2000.

M. L. Errington, T. V. Bliss, G. Richter-levin, K. Yenk, V. Doyere et al., Stimulation at 1-5 Hz does not produce long-term depression or depotentiation in the hippocampus of the adult rat in vivo, Journal of neurophysiology, vol.74, issue.4, pp.1793-1802, 1995.
URL : https://hal.archives-ouvertes.fr/hal-02540055

N. Nakai, M. Nagano, F. Saitow, Y. Watanabe, Y. Kawamura et al., Serotonin rebalances cortical tuning and behavior linked to autism symptoms in 15q11-13 CNV mice, Sci Adv, vol.3, issue.6, p.5479676, 2017.

C. Chung, S. Ha, H. Kang, J. Lee, S. M. Um et al., Early Correction of N-Methyl-D-Aspartate Receptor Function Improves Autistic-like Social Behaviors in Adult Shank2(-/-) Mice. Biological psychiatry, vol.85, pp.534-577, 2019.

P. Tovote, J. P. Fadok, and A. Luthi, Neuronal circuits for fear and anxiety, Nat Rev Neurosci, vol.16, issue.6, pp.317-348, 2015.

R. Apps and P. Strata, Neuronal circuits for fear and anxiety-the missing link, Nat Rev Neurosci, vol.16, issue.10, p.642, 2015.

G. G. Calhoon and K. M. Tye, Resolving the neural circuits of anxiety, Nat Neurosci, vol.18, issue.10, pp.1394-404, 2015.

A. Adhikari, Distributed circuits underlying anxiety, Front Behav Neurosci, vol.8, p.3978252, 2014.

E. R. Duval, A. Javanbakht, and I. Liberzon, Neural circuits in anxiety and stress disorders: a focused review, Ther Clin Risk Manag, vol.11, pp.115-141, 2015.

P. Central and P. ,

S. Duvarci, E. P. Bauer, and P. Djjon, The bed nucleus of the stria terminalis mediates inter-individual variations in anxiety and fear, vol.29, pp.10357-61, 2009.

S. Y. Kim, A. Adhikari, S. Y. Lee, J. H. Marshel, C. K. Kim et al., Diverging neural pathways assemble a behavioural state from separable features in anxiety, Nature, vol.496, issue.7444, pp.219-242, 2013.

S. N. Avery, J. A. Clauss, and J. U. Blackford, The Human BNST: Functional Role in Anxiety and Addiction, Neuropsychopharmacology, vol.41, issue.1, pp.126-167, 2016.

P. Central and P. , , p.4677124

C. Barkus, S. B. Mchugh, R. Sprengel, P. H. Seeburg, J. N. Rawlins et al., Hippocampal NMDA receptors and anxiety: at the interface between cognition and emotion, European journal of pharmacology, vol.626, issue.1, pp.49-56, 2010.

P. Central and P. ,