Browse > Article
http://dx.doi.org/10.4062/biomolther.2016.061

Clinical and Neurobiological Relevance of Current Animal Models of Autism Spectrum Disorders  

Kim, Ki Chan (Department of Neuroscience, University of Texas Southwestern Medical Center)
Gonzales, Edson Luck (Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University)
Lazaro, Maria T. (Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California)
Choi, Chang Soon (Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University)
Bahn, Geon Ho (Department of Neuropsychiatry, School of Medicine, Kyung Hee University)
Yoo, Hee Jeong (Department of Neuropsychiatry, Seoul National University Bungdang Hospital)
Shin, Chan Young (Center for Neuroscience Research, SMART Institute of Advanced Biomedical Sciences, Konkuk University)
Publication Information
Biomolecules & Therapeutics / v.24, no.3, 2016 , pp. 207-243 More about this Journal
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by social and communication impairments, as well as repetitive and restrictive behaviors. The phenotypic heterogeneity of ASD has made it overwhelmingly difficult to determine the exact etiology and pathophysiology underlying the core symptoms, which are often accompanied by comorbidities such as hyperactivity, seizures, and sensorimotor abnormalities. To our benefit, the advent of animal models has allowed us to assess and test diverse risk factors of ASD, both genetic and environmental, and measure their contribution to the manifestation of autistic symptoms. At a broader scale, rodent models have helped consolidate molecular pathways and unify the neurophysiological mechanisms underlying each one of the various etiologies. This approach will potentially enable the stratification of ASD into clinical, molecular, and neurophenotypic subgroups, further proving their translational utility. It is henceforth paramount to establish a common ground of mechanistic theories from complementing results in preclinical research. In this review, we cluster the ASD animal models into lesion and genetic models and further classify them based on the corresponding environmental, epigenetic and genetic factors. Finally, we summarize the symptoms and neuropathological highlights for each model and make critical comparisons that elucidate their clinical and neurobiological relevance.
Keywords
Autism spectrum disorders; Animal models; Genetic factors; Environmental factors; Clinical relevance;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Gauthier, J., Bonnel, A., St-Onge, J., Karemera, L., Laurent, S., Mottron, L., Fombonne, E., Joober, R. and Rouleau, G. A. (2005) NLGN3/NLGN4 gene mutations are not responsible for autism in the Quebec population. Am. J. Med. Genet. B Neuropsychiatr. Genet. 132B, 74-75.   DOI
2 Gauthier, J., Champagne, N., Lafrenière, R. G., Xiong, L., Spiegelman, D., Brustein, E., Lapointe, M., Peng, H., Côté, M., Noreau, A., Hamdan, F. F., Addington, A. M., Rapoport, J. L., Delisi, L. E., Krebs, M. O., Joober, R., Fathalli, F., Mouaffak, F., Haghighi, A. P., Néri, C., Dube, M. P., Samuels, M. E., Marineau, C., Stone, E. A., Awadalla, P., Barker, P. A., Carbonetto, S., Drapeau, P. and Rouleau, G. A. (2010) De novo mutations in the gene encoding the synaptic scaffolding protein SHANK3 in patients ascertained for schizophrenia. Proc. Natl. Acad. Sci. U.S.A. 107, 7863-7868.   DOI
3 Gepner, B. and Feron, F. (2009) Autism: a world changing too fast for a mis-wired brain? Neurosci. Biobehav. Rev. 33, 1227-1242.   DOI
4 Geschwind, D. H. (2011) Genetics of autism spectrum disorders. Trends Cogn. Sci. 15, 409-416.   DOI
5 Geschwind, D. H. and Levitt, P. (2007) Autism spectrum disorders: developmental disconnection syndromes. Curr. Opin. Neurobiol. 17, 103-111.   DOI
6 Gillberg, C. (1998) Chromosomal disorders and autism. J. Autism Dev. Disord. 28, 415-425.   DOI
7 Gillberg, C. and de Souza, L. (2002) Head circumference in autism, Asperger syndrome, and ADHD: a comparative study. Dev. Med. Child Neurol. 44, 296-300.
8 Gkogkas, C. G., Khoutorsky, A., Ran, I., Rampakakis, E., Nevarko, T., Weatherill, D. B., Vasuta, C., Yee, S., Truitt, M., Dallaire, P., Major, F., Lasko, P., Ruggero, D., Nader, K., Lacaille, J. C. and Sonenberg, N. (2013) Autism-related deficits via dysregulated eIF4Edependent translational control. Nature 493, 371-377.
9 Muroga, T., Adachi, K., Konagaya, M., Takayanagi, T. and Sobue, I. (1982) Effects of thyrotropin releasing hormone on cerebellar mutant mice--a kinesiological comparison between rolling mouse Nagoya, weaver and reeler. Jpn. J. Med. 21, 101-108.   DOI
10 Nakao, M., Sutcliffe, J. S., Durtschi, B., Mutirangura, A., Ledbetter, D. H. and Beaudet, A. L. (1994) Imprinting analysis of three genes in the Prader-Willi/Angelman region: SNRPN, E6-associated protein, and PAR-2 (D15S225E). Hum. Mol. Genet. 3, 309-315.   DOI
11 Naviaux, R. K., Zolkipli, Z., Wang, L., Nakayama, T., Naviaux, J. C., Le, T. P., Schuchbauer, M. A., Rogac, M., Tang, Q., Dugan, L. L. and Powell, S. B. (2013) Antipurinergic therapy corrects the autism-like features in the poly(IC) mouse model. PLoS One 8, e57380.   DOI
12 Need, A. C., Ge, D., Weale, M. E., Maia, J., Feng, S., Heinzen, E. L., Shianna, K. V., Yoon, W., Kasperaviciute, D., Gennarelli, M., Strittmatter, W. J., Bonvicini, C., Rossi, G., Jayathilake, K., Cola, P. A., McEvoy, J. P., Keefe, R. S., Fisher, E. M., St Jean, P. L., Giegling, I., Hartmann, A. M., Moller, H. J., Ruppert, A., Fraser, G., Crombie, C., Middleton, L. T., St Clair, D., Roses, A. D., Muglia, P., Francks, C., Rujescu, D., Meltzer, H. Y. and Goldstein, D. B. (2009) A genome-wide investigation of SNPs and CNVs in schizophrenia. PLoS Genet. 5, e1000373.   DOI
13 Nosyreva, E. D. and Huber, K. M. (2006) Metabotropic receptor-dependent long-term depression persists in the absence of protein synthesis in the mouse model of fragile X syndrome. J. Neurophysiol. 95, 3291-3295.   DOI
14 Numis, A., Major, P., Montenegro, M., Muzykewicz, D., Pulsifer, M. and Thiele, E. (2011) Identification of risk factors for autism spectrum disorders in tuberous sclerosis complex. Neurology 76, 981-987.   DOI
15 Weidner, K. L., Buenaventura, D. F. and Chadman, K. K. (2014) Mice over-expressing BDNF in forebrain neurons develop an altered behavioral phenotype with age. Behav. Brain Res. 268, 222-228.   DOI
16 Weiler, I. J., Irwin, S. A., Klintsova, A. Y., Spencer, C. M., Brazelton, A. D., Miyashiro, K., Comery, T. A., Patel, B., Eberwine, J. and Greenough, W. T. (1997) Fragile X mental retardation protein is translated near synapses in response to neurotransmitter activation. Proc. Natl. Acad. Sci. U.S.A. 94, 5395-5400.   DOI
17 Wersinger, S. R., Kelliher, K. R., Zufall, F., Lolait, S. J., O'Carroll, A. M. and Young, W. S., 3rd (2004) Social motivation is reduced in vasopressin 1b receptor null mice despite normal performance in an olfactory discrimination task. Horm. Behav. 46, 638-645.   DOI
18 Whitehouse, A. J., Bishop, D. V., Ang, Q., Pennell, C. E. and Fisher, S. E. (2011) CNTNAP2 variants affect early language development in the general population. Genes Brain and Behav. 10, 451-456.   DOI
19 Winslow, J. T. and Insel, T. R. (2002) The social deficits of the oxytocin knockout mouse. Neuropeptides 36, 221-229.   DOI
20 Wohr, M., Roullet, F. I. and Crawley, J. N. (2011a) Reduced scent marking and ultrasonic vocalizations in the BTBR T+tf/J mouse model of autism. Genes Brain Behav. 10, 35-43.   DOI
21 Wohr, M., Roullet, F. I., Hung, A. Y., Sheng, M. and Crawley, J. N. (2011b) Communication impairments in mice lacking Shank1: reduced levels of ultrasonic vocalizations and scent marking behavior. PLoS One 6, e20631.   DOI
22 Wolterink, G., Daenen, L. E., Dubbeldam, S., Gerrits, M. A., van Rijn, R., Kruse, C. G., Van Der Heijden, J. A. and Van Ree, J. M. (2001) Early amygdala damage in the rat as a model for neurodevelopmental psychopathological disorders. Eur. Neuropsychopharmacol. 11, 51-59.   DOI
23 Abraham, W. C. (2008) Metaplasticity: tuning synapses and networks for plasticity. Nat. Rev. Neurosci. 9, 387.   DOI
24 Alarcon, M., Abrahams, B. S., Stone, J. L., Duvall, J. A., Perederiy, J. V., Bomar, J. M., Sebat, J., Wigler, M., Martin, C. L., Ledbetter, D. H., Nelson, S. F., Cantor, R. M. and Geschwind, D. H. (2008) Linkage, Association, and Gene-Expression Analyses Identify CNTNAP2 as an Autism-Susceptibility Gene. Am. J. Hum. Genet. 82, 150-159.   DOI
25 Allen, G. and Courchesne, E. (2003) Differential effects of developmental cerebellar abnormality on cognitive and motor functions in the cerebellum: an fMRI study of autism. Am. J. Psychiatry 160, 262-273.   DOI
26 Altamura, C., Dell'Acqua, M. L., Moessner, R., Murphy, D. L., Lesch, K. P. and Persico, A. M. (2007) Altered neocortical cell density and layer thickness in serotonin transporter knockout mice: a quantitation study. Cereb. Cortex 17, 1394-1401.   DOI
27 Aman, M. G. and Langworthy, K. S. (2000) Pharmacotherapy for hyperactivity in children with autism and other pervasive developmental disorders. J. Autism Dev. Disord. 30, 451-459.   DOI
28 Amaral, D., Bauman, M. and Schumann, C. M. (2003) The amygdala and autism: implications from non-human primate studies. Genes Brain Behav. 2, 295-302.   DOI
29 American Psychiatric Association (2013) Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. American Psychiatric Publishing.
30 Amir, R. E., Van den Veyver, I. B., Wan, M., Tran, C. Q., Francke, U. and Zoghbi, H. Y. (1999) Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat. Genet. 23, 185-188.   DOI
31 Go, H. S., Kim, K. C., Choi, C. S., Jeon, S. J., Kwon, K. J., Han, S.-H., Lee, J., Cheong, J. H., Ryu, J. H., Kim, C.-H., Ko, K. H. and Shin, C. Y. (2012) Prenatal exposure to valproic acid increases the neural progenitor cell pool and induces macrocephaly in rat brain via a mechanism involving the GSK-$3{\beta}/{\beta}$-catenin pathway. Neuropharmacology 63, 1028-1041.   DOI
32 Goffinet, A. M. (1983) The embryonic development of the inferior olivary complex in normal and reeler (rlORL) mutant mice. J. Comp. Neurol. 219, 10-24.   DOI
33 Golan, H. M., Lev, V., Hallak, M., Sorokin, Y. and Huleihel, M. (2005) Specific neurodevelopmental damage in mice offspring following maternal inflammation during pregnancy. Neuropharmacology 48, 903-917.   DOI
34 Goorden, S. M., van Woerden, G. M., van der Weerd, L., Cheadle, J. P. and Elgersma, Y. (2007) Cognitive deficits in Tsc1+/- mice in the absence of cerebral lesions and seizures. Ann. Neurol. 62, 648-655.   DOI
35 Greer, P. L., Hanayama, R., Bloodgood, B. L., Mardinly, A. R., Lipton, D. M., Flavell, S. W., Kim, T. K., Griffith, E. C., Waldon, Z., Maehr, R., Ploegh, H. L., Chowdhury, S., Worley, P. F., Steen, J. and Greenberg, M. E. (2010) The Angelman Syndrome protein Ube3A regulates synapse development by ubiquitinating arc. Cell 140, 704-716.   DOI
36 Gregorian, C., Nakashima, J., Le Belle, J., Ohab, J., Kim, R., Liu, A., Smith, K. B., Groszer, M., Garcia, A. D., Sofroniew, M. V., Carmichael, S. T., Kornblum, H. I., Liu, X. and Wu, H. (2009) Pten deletion in adult neural stem/progenitor cells enhances constitutive neurogenesis. J. Neurosci. 29, 1874-1886.   DOI
37 Nussbaum, J., Xu, Q., Payne, T. J., Ma, J. Z., Huang, W., Gelernter, J. and Li, M. D. (2008) Significant association of the neurexin-1 gene (NRXN1) with nicotine dependence in European- and African- American smokers. Hum. Mol. Genet. 17, 1569-1577.   DOI
38 Oblak, A. L., Gibbs, T. T. and Blatt, G. J. (2011) Reduced GABAA receptors and benzodiazepine binding sites in the posterior cingulate cortex and fusiform gyrus in autism. Brain Res. 1380, 218-228.   DOI
39 Ogawa, S., Kwon, C. H., Zhou, J., Koovakkattu, D., Parada, L. F. and Sinton, C. M. (2007) A seizure-prone phenotype is associated with altered free-running rhythm in Pten mutant mice. Brain Res. 1168, 112-123.   DOI
40 Ognibene, E., Adriani, W., Macri, S. and Laviola, G. (2007) Neurobehavioural disorders in the infant reeler mouse model: interaction of genetic vulnerability and consequences of maternal separation. Behav. Brain Res. 177, 142-149.   DOI
41 Onda, H., Crino, P. B., Zhang, H., Murphey, R. D., Rastelli, L., Gould Rothberg, B. E. and Kwiatkowski, D. J. (2002) Tsc2 null murine neuroepithelial cells are a model for human tuber giant cells, and show activation of an mTOR pathway. Mol. Cell. Neurosci. 21, 561-574.   DOI
42 Ornoy, A. (2009) Valproic acid in pregnancy: how much are we endangering the embryo and fetus? Reprod. Toxicol. 28, 1-10.   DOI
43 Papaleo, F., Silverman, J. L., Aney, J., Tian, Q., Barkan, C. L., Chadman, K. K. and Crawley, J. N. (2011) Working memory deficits, increased anxiety-like traits, and seizure susceptibility in BDNF overexpressing mice. Learn. Mem. 18, 534-544.   DOI
44 Pardo, C. A. and Eberhart, C. G. (2007) The neurobiology of autism. Brain Pathol. 17, 434-447.   DOI
45 Won, H., Lee, H. R., Gee, H. Y., Mah, W., Kim, J. I., Lee, J., Ha, S., Chung, C., Jung, E. S., Cho, Y. S., Park, S. G., Lee, J. S., Lee, K., Kim, D., Bae, Y. C., Kaang, B. K., Lee, M. G. and Kim, E. (2012) Autistic-like social behaviour in Shank2-mutant mice improved by restoring NMDA receptor function. Nature 486, 261-265.   DOI
46 Wu, S., Jia, M., Ruan, Y., Liu, J., Guo, Y., Shuang, M., Gong, X., Zhang, Y., Yang, X. and Zhang, D. (2005) Positive association of the oxytocin receptor gene (OXTR) with autism in the Chinese Han population. Biol. Psychiatry 58, 74-77.   DOI
47 Yamashita, Y., Fujimoto, C., Nakajima, E., Isagai, T. and Matsuishi, T. (2003) Possible association between congenital cytomegalovirus infection and autistic disorder. J. Autism Dev. Disord. 33, 455-459.   DOI
48 Yamauchi, J., Miyamoto, Y., Kusakawa, S., Torii, T., Mizutani, R., Sanbe, A., Nakajima, H., Kiyokawa, N. and Tanoue, A. (2008) Neurofibromatosis 2 tumor suppressor, the gene induced by valproic acid, mediates neurite outgrowth through interaction with paxillin. Exp. Cell Res. 314, 2279-2288.   DOI
49 Yan, W. L., Guan, X. Y., Green, E. D., Nicolson, R., Yap, T. K., Zhang, J., Jacobsen, L. K., Krasnewich, D. M., Kumra, S., Lenane, M. C., Gochman, P., Damschroder-Williams, P. J., Esterling, L. E., Long, R. T., Martin, B. M., Sidransky, E., Rapoport, J. L. and Ginns, E. I. (2000) Childhood-onset schizophrenia/autistic disorder and t(1;7) reciprocal translocation: identification of a BAC contig spanning the translocation breakpoint at 7q21. Am. J. Med. Genet. 96, 749-753.   DOI
50 Yang, M., Bozdagi, O., Scattoni, M. L., Wöhr, M., Roullet, F. I., Katz, A. M., Abrams, D. N., Kalikhman, D., Simon, H., Woldeyohannes, L., Zhang, J. Y., Harris, M. J., Saxena, R., Silverman, J. L., Buxbaum, J. D. and Crawley, J. N. (2012) Reduced excitatory neurotransmission and mild autism-relevant phenotypes in adolescent Shank3 null mutant mice. J. Neurosci. 32, 6525-6541.   DOI
51 Amodeo, D. A., Jones, J. H., Sweeney, J. A. and Ragozzino, M. E. (2012) Differences in BTBR T+ tf/J and C57BL/6J mice on probabilistic reversal learning and stereotyped behaviors. Behav. Brain Res. 227, 64-72.   DOI
52 Andari, E., Duhamel, J. R., Zalla, T., Herbrecht, E., Leboyer, M. and Sirigu, A. (2010) Promoting social behavior with oxytocin in highfunctioning autism spectrum disorders. Proc. Natl. Acad. Sci. U.S.A. 107, 4389-4394.   DOI
53 Arking, D. E., Cutler, D. J., Brune, C. W., Teslovich, T. M., West, K., Ikeda, M., Rea, A., Guy, M., Lin, S., Cook E. H. and Chakravarti, A. (2008) A common genetic variant in the neurexin superfamily member CNTNAP2 increases familial risk of autism. Am. J. Hum. Genet. 82, 160-164.   DOI
54 Arndt, T. L., Stodgell, C. J. and Rodier, P. M. (2005) The teratology of autism. Int. J. Dev. Neurosci. 23, 189-199.   DOI
55 Aronson, M., Hagberg, B. and Gillberg, C. (1997) Attention deficits and autistic spectrum problems in children exposed to alcohol during gestation: a follow-up study. Dev. Med. Child Neurol. 39, 583-587.
56 Ashwood, P., Krakowiak, P., Hertz-Picciotto, I., Hansen, R., Pessah, I. and Van de Water, J. (2011) Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav. Immun. 25, 40-45.   DOI
57 Auerbach, B. D., Osterweil, E. K. and Bear, M. F. (2011) Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature 480, 63-68.   DOI
58 Autism and Developmental Disabilities Monitoring Network Surveillance Year 2010 Principal Investigators (2014) Prevalence of autism spectrum disorder among children aged 8 years-autism and developmental disabilities monitoring network, 11 sites, United States, 2010. MMWR Surveill. Summ. 63, 1-21.
59 Gregory, S. G., Connelly, J. J., Towers, A. J., Johnson, J., Biscocho, D., Markunas, C. A., Lintas, C., Abramson, R. K., Wright, H. H., Ellis, P., Langford, C. F., Worley, G., Delong, G. R., Murphy, S. K., Cuccaro, M. L., Persico, A. and Pericak-Vance, M. A. (2009) Genomic and epigenetic evidence for oxytocin receptor deficiency in autism. BMC Med. 7, 62.   DOI
60 Groc, L., Choquet, D., Stephenson, F. A., Verrier, D., Manzoni, O. J. and Chavis, P. (2007) NMDA receptor surface trafficking and synaptic subunit composition are developmentally regulated by the extracellular matrix protein Reelin. J. Neurosci. 27, 10165-10175.   DOI
61 Guastella, A. J., Einfeld, S. L., Gray, K. M., Rinehart, N. J., Tonge, B. J., Lambert, T. J. and Hickie, I. B. (2010) Intranasal oxytocin improves emotion recognition for youth with autism spectrum disorders. Biol. Psychiatry 67, 692-694.   DOI
62 Guastella, A. J., Mitchell, P. B. and Dadds, M. R. (2008) Oxytocin increases gaze to the eye region of human faces. Biol. Psychiatry 63, 3-5.   DOI
63 Guffanti, G., Lievers, L. S., Bonati, M. T., Marchi, M., Geronazzo, L., Nardocci, N., Estienne, M., Larizza, L., Macciardi, F. and Russo, S. (2011) Role of UBE3A and ATP10A genes in autism susceptibility region 15q11-q13 in an Italian population: A positive replication for UBE3A. Psychiatry Res. 185, 33-38.   DOI
64 Guy, J., Cheval, H., Selfridge, J. and Bird, A. (2011) The Role of MeCP2 in the Brain. Annu. Rev. Cell Dev. Biol. 27, 631-652.   DOI
65 Guy, J., Hendrich, B., Holmes, M., Martin, J. E. and Bird, A. (2001) A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat. Genet. 27, 322-326.   DOI
66 Parker, K. J., Garner, J. P., Libove, R. A., Hyde, S. A., Hornbeak, K. B., Carson, D. S., Liao, C. P., Phillips, J. M., Hallmayer, J. F. and Hardan, A. Y. (2014) Plasma oxytocin concentrations and OXTR polymorphisms predict social impairments in children with and without autism spectrum disorder. Proc. Natl. Acad. Sci. U.S.A. 111, 12258-12263.   DOI
67 Patrylo, P. R., Browning, R. A. and Cranick, S. (2006) Reeler homozygous mice exhibit enhanced susceptibility to epileptiform activity. Epilepsia 47, 257-266.
68 Patterson, P. H. (2009) Immune involvement in schizophrenia and autism: etiology, pathology and animal models. Behav. Brain Res. 204, 313-321.   DOI
69 Patterson, P. H. (2011) Maternal infection and immune involvement in autism. Trends Mol. Med. 17, 389-394.   DOI
70 Peca, J., Feliciano, C., Ting, J. T., Wang, W., Wells, M. F., Venkatraman, T. N., Lascola, C. D., Fu, Z. and Feng, G. (2011) Shank3 mutant mice display autistic-like behaviours and striatal dysfunction. Nature 472, 437-442.   DOI
71 Penagarikano, O., Abrahams, B. S., Herman, E. I., Winden, K. D., Gdalyahu, A., Dong, H., Sonnenblick, L. I., Gruver, R., Almajano, J., Bragin, A., Golshani, P., Trachtenberg, J. T., Peles, E. and Geschwind, D. H. (2011) Absence of CNTNAP2 Leads to Epilepsy, Neuronal Migration Abnormalities, and Core Autism-Related Deficits. Cell 147, 235-246.   DOI
72 Penagarikano, O., Lazaro, M. T., Lu, X. H., Gordon, A., Dong, H., Lam, H. A., Peles, E., Maidment, N. T., Murphy, N. P., Yang, X. W., Golshani, P. and Geschwind, D. H. (2015) Exogenous and evoked oxytocin restores social behavior in the Cntnap2 mouse model of autism. Sci. Transl. Med. 7, 271ra8.   DOI
73 Yasui, D. H., Peddada, S., Bieda, M. C., Vallero, R. O., Hogart, A., Nagarajan, R. P., Thatcher, K. N., Farnham, P. J. and Lasalle, J. M. (2007) Integrated epigenomic analyses of neuronal MeCP2 reveal a role for long-range interaction with active genes. Proc. Natl. Acad. Sci. U.S.A. 104, 19416-19421.   DOI
74 Yip, J. W., Yip, Y. P., Nakajima, K. and Capriotti, C. (2000) Reelin controls position of autonomic neurons in the spinal cord. Proc. Natl. Acad. Sci. U.S.A. 97, 8612-8616.   DOI
75 Yirmiya, N., Rosenberg, C., Levi, S., Salomon, S., Shulman, C., Nemanov, L., Dina, C. and Ebstein, R. P. (2006) Association between the arginine vasopressin 1a receptor (AVPR1a) gene and autism in a family-based study: mediation by socialization skills. Mol. Psychiatry 11, 488-494.   DOI
76 Ylisaukko-oja, T., Rehnström, K., Auranen, M., Vanhala, R., Alen, R., Kempas, E., Ellonen, P., Turunen, J. A., Makkonen, I., Riikonen, R., Nieminen-von Wendt, T., von Wendt, L., Peltonen, L. and Jarvela, I. (2005) Analysis of four neuroligin genes as candidates for autism. Eur. J. Hum. Genet. 13, 1285-1292.   DOI
77 Young, D. M., Schenk, A. K., Yang, S. B., Jan, Y. N. and Jan, L. Y. (2010) Altered ultrasonic vocalizations in a tuberous sclerosis mouse model of autism. Proc. Natl. Acad. Sci. U.S.A. 107, 11074-11079.   DOI
78 Zec, N., Rowitch, D. H., Bitgood, M. J. and Kinney, H. C. (1997) Expression of the homeobox-containing genes EN1 and EN2 in human fetal midgestational medulla and cerebellum. J. Neuropathol. Exp. Neurol. 56, 236-242.   DOI
79 Zhang, J., Hou, L., Klann, E. and Nelson, D. L. (2009) Altered hippocampal synaptic plasticity in the FMR1 gene family knockout mouse models. J. Neurophysiol. 101, 2572-2580.   DOI
80 Bachevalier, J. (1994) Medial temporal lobe structures and autism: a review of clinical and experimental findings. Neuropsychologia 32, 627-648.   DOI
81 Bailey, A., Le Couteur, A., Gottesman, I., Bolton, P., Simonoff, E., Yuzda, E. and Rutter, M. (1995) Autism as a strongly genetic disorder: evidence from a British twin study. Psychol. Med. 25, 63-77.   DOI
82 Ballas, N., Lioy, D. T., Grunseich, C. and Mandel, G. (2009) Non-cell autonomous influence of MeCP2-deficient glia on neuronal dendritic morphology. Nat. Neurosci. 12, 311-317.   DOI
83 Barnea-Goraly, N., Frazier, T. W., Piacenza, L., Minshew, N. J., Keshavan, M. S., Reiss, A. L. and Hardan, A. Y. (2014) A preliminary longitudinal volumetric MRI study of amygdala and hippocampal volumes in autism. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 48, 124-128.   DOI
84 Bateup, H. S., Takasaki, K. T., Saulnier, J. L., Denefrio, C. L. and Sabatini, B. L. (2011) Loss of Tsc1 in vivo impairs hippocampal mGluRLTD and increases excitatory synaptic function. J. Neurosci. 31, 8862-8869.   DOI
85 Bauman, M. and Kemper, T. L. (1985) Histoanatomic observations of the brain in early infantile autism. Neurology 35, 866-874.   DOI
86 Bear, M. F., Huber, K. M. and Warren, S. T. (2004) The mGluR theory of fragile X mental retardation. Trends Neurosci. 27, 370-377.   DOI
87 Bello, S. C. (2007) Autism and environmental influences: review and commentary. Rev. Environ. Health 22, 139-156.
88 Hallmayer, J., Cleveland, S., Torres, A., Phillips, J., Cohen, B., Torigoe, T., Miller, J., Fedele, A., Collins, J., Smith, K., Lotspeich, L., Croen, L. A., Ozonoff, S., Lajonchere, C., Grether, J. K. and Risch, N. (2011) Genetic Heritability and Shared Environmental Factors Among Twin Pairs With Autism. Arch. Gen. Psychiatry 68, 1095-1102.   DOI
89 Hardan, A. Y., Minshew, N. J., Mallikarjuhn, M. and Keshavan, M. S. (2001) Brain volume in autism. J. Child Neurol. 16, 421-424.   DOI
90 Hatton, D. D., Sideris, J., Skinner, M., Mankowski, J., Bailey, D. B., Jr., Roberts, J. and Mirrett, P. (2006) Autistic behavior in children with fragile X syndrome: prevalence, stability, and the impact of FMRP. Am. J. Med. Genet. A 140A, 1804-1813.   DOI
91 Hava, G., Vered, L., Yael, M., Mordechai, H. and Mahoud, H. (2006) Alterations in behavior in adult offspring mice following maternal inflammation during pregnancy. Dev. Psychobiol. 48, 162-168.   DOI
92 Hazlett, H. C., Poe, M., Gerig, G., Smith, R. G., Provenzale, J., Ross, A., Gilmore, J. and Piven, J. (2005) Magnetic resonance imaging and head circumference study of brain size in autism: birth through age 2 years. Arch. Gen. Psychiatry 62, 1366-1376.   DOI
93 Hertz-Picciotto, I., Croen, L. A., Hansen, R., Jones, C. R., van de Water, J. and Pessah, I. N. (2006) The CHARGE study: an epidemiologic investigation of genetic and environmental factors contributing to autism. Environ. Health Perspect. 114, 1119-1125.   DOI
94 Hoeffer, C. A., Santini, E., Ma, T., Arnold, E. C., Whelan, A. M., Wong, H., Pierre, P., Pelletier, J. and Klann, E. (2013) Multiple components of eIF4F are required for protein synthesis-dependent hippocampal long-term potentiation. J. Neurophysiol. 109, 68-76.   DOI
95 Perry, E. K., Lee, M. L., Martin-Ruiz, C. M., Court, J. A., Volsen, S. G., Merrit, J., Folly, E., Iversen, P. E., Bauman, M. L., Perry, R. H. and Wenk, G. L. (2001) Cholinergic activity in autism: abnormalities in the cerebral cortex and basal forebrain. Am. J. Psychiatry 158, 1058-1066.   DOI
96 Peters, S., Beaudet, A., Madduri, N. and Bacino, C. (2004) Autism in Angelman syndrome: implications for autism research. Clin. Genet. 66, 530-536.   DOI
97 Pierce, K. and Courchesne, E. (2001) Evidence for a cerebellar role in reduced exploration and stereotyped behavior in autism. Biol. Psychiatry 49, 655-664.   DOI
98 Pletnikov, M. V., Moran, T. H. and Carbone, K. M. (2002) Borna disease virus infection of the neonatal rat: developmental brain injury model of autism spectrum disorders. Front. Biosci. 7, d593-d607.
99 Pletnikov, M. V., Rubin, S. A., Vasudevan, K., Moran, T. H. and Carbone, K. M. (1999) Developmental brain injury associated with abnormal play behavior in neonatally Borna disease virus-infected Lewis rats: a model of autism. Behav. Brain Res. 100, 43-50.   DOI
100 Pobbe, R. L., Pearson, B. L., Defensor, E. B., Bolivar, V. J., Young, W. S., 3rd, Lee, H. J., Blanchard, D. C. and Blanchard, R. J. (2012) Oxytocin receptor knockout mice display deficits in the expression of autism-related behaviors. Horm. Behav. 61, 436-444.   DOI
101 Podhorna, J. and Didriksen, M. (2004) The heterozygous reeler mouse: behavioural phenotype. Behav. Brain Res. 153, 43-54.   DOI
102 Poliak, S., Gollan, L., Martinez, R., Custer, A., Einheber, S., Salzer, J. L., Trimmer, J. S., Shrager, P. and Peles, E. (1999) Caspr2, a New Member of the Neurexin Superfamily, Is Localized at the Juxtaparanodes of Myelinated Axons and Associates with K+ Channels. Neuron 24, 1037-1047.   DOI
103 Zhong, H., Serajee, F. J., Nabi, R. and Huq, A. H. (2003) No association between the EN2 gene and autistic disorder. J. Med. Genet. 40, e4.   DOI
104 Zhou, J. and Parada, L. F. (2012) PTEN signaling in autism spectrum disorders. Curr. Opin. Neurobiol. 22, 873-879.   DOI
105 Benayed, R., Gharani, N., Rossman, I., Mancuso, V., Lazar, G., Kamdar, S., Bruse, S. E., Tischfield, S., Smith, B. J., Zimmerman, R. A., Dicicco-Bloom, E., Brzustowicz, L. M. and Millonig, J. H. (2005) Support for the homeobox transcription factor gene ENGRAILED 2 as an autism spectrum disorder susceptibility locus. Am. J. Hum. Genet. 77, 851-868.   DOI
106 Benvenuto, A., Moavero, R., Alessandrelli, R., Manzi, B. and Curatolo, P. (2009) Syndromic autism: causes and pathogenetic pathways. World J. Pediatr. 5, 169-176.   DOI
107 Berkel, S., Marshall, C. R., Weiss, B., Howe, J., Roeth, R., Moog, U., Endris, V., Roberts, W., Szatmari, P., Pinto, D., Bonin, M., Riess, A., Engels, H., Sprengel, R., Scherer, S. W. and Rappold, G. A. (2010) Mutations in the SHANK2 synaptic scaffolding gene in autism spectrum disorder and mental retardation. Nat. Genet. 42, 489-491.   DOI
108 Berkel, S., Tang, W., Treviño, M., Vogt, M., Obenhaus, H. A., Gass, P., Scherer, S. W., Sprengel, R., Schratt, G. and Rappold, G. A. (2012) Inherited and de novo SHANK2 variants associated with autism spectrum disorder impair neuronal morphogenesis and physiology. Hum. Mol. Genet. 21, 344-357.   DOI
109 Bernard, S., Enayati, A., Roger, H., Binstock, T. and Redwood, L. (2002) The role of mercury in the pathogenesis of autism. Mol. Psychiatry 7 Suppl 2, S42-S43.   DOI
110 Bernardet, M. and Crusio, W. E. (2006) Fmr1 KO mice as a possible model of autistic features. Sci. World J. 6, 1164-1176.   DOI
111 Betancur, C., Corbex, M., Spielewoy, C., Philippe, A., Laplanche, J. L., Launay, J. M., Gillberg, C., Mouren-Siméoni, M. C., Hamon, M., Giros, B., Nosten-Bertrand, M. and Leboyer, M. (2002) Serotonin transporter gene polymorphisms and hyperserotonemia in autistic disorder. Mol. Psychiatry 7, 67-71.   DOI
112 Hollander, E., Novotny, S., Hanratty, M., Yaffe, R., DeCaria, C. M., Aronowitz, B. R. and Mosovich, S. (2003) Oxytocin infusion reduces repetitive behaviors in adults with autistic and Asperger's disorders. Neuropsychopharmacology 28, 193-198.   DOI
113 Holmes, A., Yang, R. J., Lesch, K. P., Crawley, J. N. and Murphy, D. L. (2003) Mice lacking the serotonin transporter exhibit 5-HT(1A) receptor-mediated abnormalities in tests for anxiety-like behavior. Neuropsychopharmacology 28, 2077-2088.   DOI
114 Homanics, G. E., DeLorey, T. M., Firestone, L. L., Quinlan, J. J., Handforth, A., Harrison, N. L., Krasowski, M. D., Rick, C. E., Korpi, E. R., Makela, R., Brilliant, M. H., Hagiwara, N., Ferguson, C., Snyder, K. and Olsen, R. W. (1997) Mice devoid of gamma-aminobutyrate type A receptor beta3 subunit have epilepsy, cleft palate, and hypersensitive behavior. Proc. Natl. Acad. Sci. U.S.A. 94, 4143-4148.   DOI
115 Hornig, M., Weissenbock, H., Horscroft, N. and Lipkin, W. I. (1999) An infection-based model of neurodevelopmental damage. Proc. Natl. Acad. Sci. U.S.A. 96, 12102-12107.   DOI
116 Hranilovic, D., Novak, R., Babic, M., Novokmet, M., Bujas-Petkovic, Z. and Jernej, B. (2008) Hyperserotonemia in autism: the potential role of 5HT-related gene variants. Coll. Antropol. 32 Suppl 1, 75-80.
117 Hsiao, E. Y., McBride, S. W., Chow, J., Mazmanian, S. K. and Patterson, P. H. (2012) Modeling an autism risk factor in mice leads to permanent immune dysregulation. Proc. Natl. Acad. Sci. U.S.A. 109, 12776-12781.   DOI
118 Huang, C. H. and Santangelo, S. L. (2008) Autism and serotonin transporter gene polymorphisms: A systematic review and meta-analysis. Am. J. Med. Genet. B Neuropsychiatr. Genet. 147B, 903-913.   DOI
119 Popova, N. K., Vishnivetskaya, G. B., Ivanova, E. A., Skrinskaya, J. A. and Seif, I. (2000) Altered behavior and alcohol tolerance in transgenic mice lacking MAO A: a comparison with effects of MAO A inhibitor clorgyline. Pharmacol. Biochem. Behav. 67, 719-727.   DOI
120 Prasad, H. C., Steiner, J. A., Sutcliffe, J. S. and Blakely, R. D. (2009) Enhanced activity of human serotonin transporter variants associated with autism. Philos. Trans. R. Soc. Lond., B, Biol. Sci. 364, 163-173.   DOI
121 Radyushkin, K., Hammerschmidt, K., Boretius, S., Varoqueaux, F., El- Kordi, A., Ronnenberg, A., Winter, D., Frahm, J., Fischer, J., Brose, N. and Ehrenreich, H. (2009) Neuroligin-3-deficient mice: model of a monogenic heritable form of autism with an olfactory deficit. Genes Brain Behav. 8, 416-425.   DOI
122 Rasmussen, S. A. and Friedman, J. M. (2000) NF1 gene and neurofibromatosis 1. Am. J. Epidemiol. 151, 33-40.   DOI
123 Reith, R. M., Way, S., McKenna, J., 3rd, Haines, K. and Gambello, M. J. (2011) Loss of the tuberous sclerosis complex protein tuberin causes Purkinje cell degeneration. Neurobiol. Dis. 43, 113-122.   DOI
124 Rett, A. (1966) [On a unusual brain atrophy syndrome in hyperammonemia in childhood]. Wien. Med. Wochenschr. 116, 723-726.
125 Richt, J. A., Pfeuffer, I., Christ, M., Frese, K., Bechter, K. and Herzog, S. (1997) Borna disease virus infection in animals and humans. Emerging Infect. Dis. 3, 343-352.   DOI
126 Rinaldi, T., Kulangara, K., Antoniello, K. and Markram, H. (2007) Elevated NMDA receptor levels and enhanced postsynaptic long-term potentiation induced by prenatal exposure to valproic acid. Proc. Natl. Acad. Sci. U.S.A. 104, 13501-13506.   DOI
127 Bielsky, I. F., Hu, S. B., Ren, X., Terwilliger, E. F. and Young, L. J. (2005) The V1a vasopressin receptor is necessary and sufficient for normal social recognition: a gene replacement study. Neuron 47, 503-513.   DOI
128 Bill, B. R. and Geschwind, D. H. (2009) Genetic advances in autism: heterogeneity and convergence on shared pathways. Curr. Opin. Genet. Dev. 19, 271-278.   DOI
129 Binder, D. K. and Scharfman, H. E. (2004) Brain-derived neurotrophic factor. Growth Factors 22, 123-131.   DOI
130 Blanchard, D. C., Defensor, E. B., Meyza, K. Z., Pobbe, R. L., Pearson, B. L., Bolivar, V. J. and Blanchard, R. J. (2012) BTBR T+tf/J mice: autism-relevant behaviors and reduced fractone-associated heparan sulfate. Neurosci. Biobehav. Rev. 36, 285-296.   DOI
131 Blatt, G. J. (2005) GABAergic cerebellar system in autism: a neuropathological and developmental perspective. Int. Rev. Neurobiol. 71, 167-178.   DOI
132 Blatt, G. J., Fitzgerald, C. M., Guptill, J. T., Booker, A. B., Kemper, T. L. and Bauman, M. L. (2001) Density and distribution of hippocampal neurotransmitter receptors in autism: an autoradiographic study. J. Autism Dev. Disord. 31, 537-543.   DOI
133 Blundell, J., Blaiss, C. A., Etherton, M. R., Espinosa, F., Tabuchi, K., Walz, C., Bolliger, M. F., Sudhof, T. C. and Powell, C. M. (2010) Neuroligin-1 deletion results in impaired spatial memory and increased repetitive behavior. J. Neurosci. 30, 2115-2129.   DOI
134 Bobee, S., Mariette, E., Tremblay-Leveau, H. and Caston, J. (2000) Effects of early midline cerebellar lesion on cognitive and emotional functions in the rat. Behav. Brain Res. 112, 107-117.   DOI
135 Huang, E. J. and Reichardt, L. F. (2001) Neurotrophins: roles in neuronal development and function. Annu. Rev. Neurosci. 24, 677-736.   DOI
136 Hung, A. Y., Futai, K., Sala, C., Valtschanoff, J. G., Ryu, J., Woodworth, M. A., Kidd, F. L., Sung, C. C., Miyakawa, T., Bear, M. F., Weinberg, R. J. and Sheng, M. (2008) Smaller dendritic spines, weaker synaptic transmission, but enhanced spatial learning in mice lacking Shank1. J. Neurosci. 28, 1697-1708.   DOI
137 Hunter, P. (2010) The psycho gene. EMBO Rep. 11, 667-669.   DOI
138 Husi, H., Ward, M. A., Choudhary, J. S., Blackstock, W. P. and Grant, S. G. (2000) Proteomic analysis of NMDA receptor-adhesion protein signaling complexes. Nat. Neurosci. 3, 661-669.   DOI
139 Hutsler, J. J. and Zhang, H. (2010) Increased dendritic spine densities on cortical projection neurons in autism spectrum disorders. Brain Res. 1309, 83-94.   DOI
140 Iafrati, J., Orejarena, M., Lassalle, O., Bouamrane, L., Gonzalez- Campo, C. and Chavis, P. (2014) Reelin, an extracellular matrix protein linked to early onset psychiatric diseases, drives postnatal development of the prefrontal cortex via GluN2B-NMDARs and the mTOR pathway. Mol. Psychiatry 19, 417-426.   DOI
141 Insel, T. R. (2010) The challenge of translation in social neuroscience: a review of oxytocin, vasopressin, and affiliative behavior. Neuron 65, 768-779.   DOI
142 Irons, M., Elias, E. R., Salen, G., Tint, G. S. and Batta, A. K. (1993) Defective cholesterol biosynthesis in Smith-Lemli-Opitz syndrome. Lancet 341, 1414.
143 Irwin, S. A., Galvez, R. and Greenough, W. T. (2000) Dendritic spine structural anomalies in fragile-X mental retardation syndrome. Cereb. Cortex 10, 1038-1044.   DOI
144 Rinaldi, T., Perrodin, C. and Markram, H. (2008) Hyper-connectivity and hyper-plasticity in the medial prefrontal cortex in the valproic Acid animal model of autism. Front. Neural Circuits 2, 4.
145 Rios, M., Lambe, E. K., Liu, R., Teillon, S., Liu, J., Akbarian, S., Roffler- Tarlov, S., Jaenisch, R. and Aghajanian, G. K. (2006) Severe deficits in 5-HT2A -mediated neurotransmission in BDNF conditional mutant mice. J. Neurobiol. 66, 408-420.   DOI
146 Rogers, J. T., Rusiana, I., Trotter, J., Zhao, L., Donaldson, E., Pak, D. T., Babus, L. W., Peters, M., Banko, J. L., Chavis, P., Rebeck, G. W., Hoe, H. S. and Weeber, E. J. (2011) Reelin supplementation enhances cognitive ability, synaptic plasticity, and dendritic spine density. Learn. Mem. 18, 558-564.   DOI
147 Rogers, S. J., Hepburn, S. and Wehner, E. (2003) Parent reports of sensory symptoms in toddlers with autism and those with other developmental disorders. J. Autism Dev. Disord. 33, 631-642.   DOI
148 Romero, E., Ali, C., Molina-Holgado, E., Castellano, B., Guaza, C. and Borrell, J. (2007) Neurobehavioral and immunological consequences of prenatal immune activation in rats. Influence of antipsychotics. Neuropsychopharmacology 32, 1791-1804.   DOI
149 Rout, U. K. and Dhossche, D. M. (2008) A pathogenetic model of autism involving Purkinje cell loss through anti-GAD antibodies. Med. Hypotheses 71, 218-221.   DOI
150 Rutter, M., Caspi, A. and Moffitt, T. E. (2003) Using sex differences in psychopathology to study causal mechanisms: unifying issues and research strategies. J. Child Psychol. Psychiatry 44, 1092-1115.   DOI
151 Boccaccio, I., Glatt-Deeley, H., Watrin, F., Roeckel, N., Lalande, M. and Muscatelli, F. (1999) The human MAGEL2 gene and its mouse homologue are paternally expressed and mapped to the Prader- Willi region. Hum. Mol. Genet. 8, 2497-2505.   DOI
152 Bolivar, V. J., Walters, S. R. and Phoenix, J. L. (2007) Assessing autism- like behavior in mice: variations in social interactions among inbred strains. Behav. Brain Res. 176, 21-26.   DOI
153 Bolton, P. F., Park, R. J., Higgins, J. N., Griffiths, P. D. and Pickles, A. (2002) Neuro-epileptic determinants of autism spectrum disorders in tuberous sclerosis complex. Brain 125, 1247-1255.   DOI
154 Bonaguidi, M. A., Wheeler, M. A., Shapiro, J. S., Stadel, R. P., Sun, G. J., Ming, G. L. and Song, H. (2011) In vivo clonal analysis reveals self-renewing and multipotent adult neural stem cell characteristics. Cell 145, 1142-1155.   DOI
155 Bortolato, M., Godar, S. C., Alzghoul, L., Zhang, J., Darling, R. D., Simpson, K. L., Bini, V., Chen, K., Wellman, C. L., Lin, R. C. and Shih, J. C. (2013) Monoamine oxidase A and A/B knockout mice display autistic-like features. Int. J. Neuropsychopharmacol. 16, 869-888.   DOI
156 Bozdagi, O., Sakurai, T., Papapetrou, D., Wang, X., Dickstein, D. L., Takahashi, N., Kajiwara, Y., Yang, M., Katz, A. M., Scattoni, M. L., Harris, M. J., Saxena, R., Silverman, J. L., Crawley, J. N., Zhou, Q., Hof, P. R. and Buxbaum, J. D. (2010) Haploinsufficiency of the autism- associated Shank3 gene leads to deficits in synaptic function, social interaction, and social communication. Mol. Autism 1, 15.   DOI
157 Brennan, F. X., Albeck, D. S. and Paylor, R. (2006) Fmr1 knockout mice are impaired in a leverpress escape/avoidance task. Genes Brain Behav. 5, 467-471.   DOI
158 Jacob, S., Brune, C. W., Carter, C. S., Leventhal, B. L., Lord, C. and Cook, E. H., Jr. (2007) Association of the oxytocin receptor gene (OXTR) in Caucasian children and adolescents with autism. Neurosci Lett. 417, 6-9.   DOI
159 Jamain, S., Quach, H., Betancur, C., Rastam, M., Colineaux, C., Gillberg, I. C., Soderstrom, H., Giros, B., Leboyer, M., Gillberg, C. and Bourgeron, T. (2003) Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nat. Genet. 34, 27-29.   DOI
160 Jamain, S., Radyushkin, K., Hammerschmidt, K., Granon, S., Boretius, S., Varoqueaux, F., Ramanantsoa, N., Gallego, J., Ronnenberg, A., Winter, D., Frahm, J., Fischer, J., Bourgeron, T., Ehrenreich, H. and Brose, N. (2008) Reduced social interaction and ultrasonic communication in a mouse model of monogenic heritable autism. Proc. Natl. Acad. Sci. U.S.A. 105, 1710-1715.   DOI
161 Jiang, X., Wang, J., Luo, T. and Li, Q. (2009) Impaired hypothalamicpituitary- adrenal axis and its feedback regulation in serotonin transporter knockout mice. Psychoneuroendocrinology 34, 317-331.   DOI
162 Jiang, Y. H. and Ehlers, M. D. (2013) Modeling Autism by SHANK Gene Mutations in Mice. Neuron 78, 8-27.   DOI
163 Jiang, Y. H., Pan, Y., Zhu, L., Landa, L., Yoo, J., Spencer, C., Lorenzo, I., Brilliant, M., Noebels, J. and Beaudet, A. L. (2010) Altered ultrasonic vocalization and impaired learning and memory in Angelman syndrome mouse model with a large maternal deletion from Ube3a to Gabrb3. PLoS One 5, e12278.   DOI
164 Jiang, Y. H., Sahoo, T., Michaelis, R. C., Bercovich, D., Bressler, J., Kashork, C. D., Liu, Q., Shaffer, L. G., Schroer, R. J., Stockton, D. W., Spielman, R. S., Stevenson, R. E. and Beaudet, A. L. (2004) A mixed epigenetic/genetic model for oligogenic inheritance of autism with a limited role for UBE3A. Am. J. Med. Genet. A 131, 1-10.
165 Sala, M., Braida, D., Lentini, D., Busnelli, M., Bulgheroni, E., Capurro, V., Finardi, A., Donzelli, A., Pattini, L., Rubino, T., Parolaro, D., Nishimori, K., Parenti, M. and Chini, B. (2011) Pharmacologic rescue of impaired cognitive flexibility, social deficits, increased aggression, and seizure susceptibility in oxytocin receptor null mice: a neurobehavioral model of autism. Biol. Psychiatry 69, 875-882.   DOI
166 Salinger, W. L., Ladrow, P. and Wheeler, C. (2003) Behavioral phenotype of the reeler mutant mouse: effects of RELN gene dosage and social isolation. Behav. Neurosci. 117, 1257-1275.   DOI
167 Samaco, R. C., Hogart, A. and LaSalle, J. M. (2005) Epigenetic overlap in autism-spectrum neurodevelopmental disorders: MECP2 deficiency causes reduced expression of UBE3A and GABRB3. Hum. Mol. Genet. 14, 483-492.   DOI
168 Sampath, S., Bhat, S., Gupta, S., O'Connor, A., West, A. B., Arking, D. E. and Chakravarti, A. (2013) Defining the Contribution of CNTNAP2 to Autism Susceptibility. PLoS One 8, e77906.   DOI
169 Santini, E., Huynh, T. N., MacAskill, A. F., Carter, A. G., Pierre, P., Ruggero, D., Kaphzan, H. and Klann, E. (2013) Exaggerated translation causes synaptic and behavioural aberrations associated with autism. Nature 493, 411-415.
170 Sato, D., Lionel, A. C., Leblond, C. S., Prasad, A., Pinto, D., Walker, S., O'Connor, I., Russell, C., Drmic, I. E., Hamdan, F. F., Michaud, J. L., Endris, V., Roeth, R., Delorme, R., Huguet, G., Leboyer, M., Rastam, M., Gillberg, C., Lathrop, M., Stavropoulos, D. J., Anagnostou, E., Weksberg, R., Fombonne, E., Zwaigenbaum, L., Fernandez, B. A., Roberts, W., Rappold, G. A., Marshall, C. R., Bourgeron, T., Szatmari, P. and Scherer, S. W. (2012) SHANK1 Deletions in Males with Autism Spectrum Disorder. Am. J. Hum. Genet. 90, 879-887.   DOI
171 Brune, C. W., Korvatska, E., Allen-Brady, K., Cook, E. H., Jr., Dawson, G., Devlin, B., Estes, A., Hennelly, M., Hyman, S. L., McMahon, W. M., Munson, J., Rodier, P. M., Schellenberg, G. D., Stodgell, C. J. and Coon, H. (2008) Heterogeneous association between engrailed- 2 and autism in the CPEA network. Am. J. Med. Genet. B Neuropsychiatr. Genet. 147B, 187-193.   DOI
172 Bryn, V., Halvorsen, B., Ueland, T., Isaksen, J., Kolkova, K., Ravn, K. and Skjeldal, O. (2015) Brain derived neurotrophic factor (BDNF) and autism spectrum disorders (ASD) in childhood. Eur. J. Paediatr. Neurol. 19, 411-414.   DOI
173 Busquets-Garcia, A., Gomis-González, M., Guegan, T., Agustín- Pavon, C., Pastor, A., Mato, S., Pérez-Samartín, A., Matute, C., de la Torre, R., Dierssen, M., Maldonado, R. and Ozaita, A. (2013) Targeting the endocannabinoid system in the treatment of fragile X syndrome. Nat. Med. 19, 603-607.   DOI
174 Butler, M. G., Dasouki, M. J., Zhou, X. P., Talebizadeh, Z., Brown, M., Takahashi, T. N., Miles, J. H., Wang, C. H., Stratton, R., Pilarski, R. and Eng, C. (2005) Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations. J. Med. Genet. 42, 318-321.   DOI
175 Buxbaum, J. D., Silverman, J. M., Smith, C. J., Greenberg, D. A., Kilifarski, M., Reichert, J., Cook, E. H., Jr., Fang, Y., Song, C. Y. and Vitale, R. (2002) Association between a GABRB3 polymorphism and autism. Mol. Psychiatry 7, 311-316.   DOI
176 Caldwell, H. K., Wersinger, S. R. and Young, W. S., 3rd (2008) The role of the vasopressin 1b receptor in aggression and other social behaviours. Prog. Brain Res. 170, 65-72.   DOI
177 Carper, R. A. and Courchesne, E. (2005) Localized enlargement of the frontal cortex in early autism. Biol. Psychiatry 57, 126-133.   DOI
178 Joyal, C. C., Meyer, C., Jacquart, G., Mahler, P., Caston, J. and Lalonde, R. (1996) Effects of midline and lateral cerebellar lesions on motor coordination and spatial orientation. Brain Res. 739, 1-11.   DOI
179 Jung, G. A., Yoon, J. Y., Moon, B. S., Yang, D. H., Kim, H. Y., Lee, S. H., Bryja, V., Arenas, E. and Choi, K. Y. (2008) Valproic acid induces differentiation and inhibition of proliferation in neural progenitor cells via the beta-catenin-Ras-ERK-p21Cip/WAF1 pathway. BMC Cell Biol. 9, 66.   DOI
180 Jung, K.-M., Sepers, M., Henstridge, C. M., Lassalle, O., Neuhofer, D., Martin, H., Ginger, M., Frick, A., DiPatrizio, N. V., Mackie, K., Katona, I., Piomelli, D. and Manzoni, O. J. (2012) Uncoupling of the endocannabinoid signalling complex in a mouse model of fragile X syndrome. Nat. Commun. 3, 1080.   DOI
181 Jyonouchi, H., Geng, L., Ruby, A. and Zimmerman-Bier, B. (2005) Dysregulated innate immune responses in young children with autism spectrum disorders: their relationship to gastrointestinal symptoms and dietary intervention. Neuropsychobiology 51, 77-85.   DOI
182 Kalueff, A. V., Fox, M. A., Gallagher, P. S. and Murphy, D. L. (2007) Hypolocomotion, anxiety and serotonin syndrome-like behavior contribute to the complex phenotype of serotonin transporter knockout mice. Genes Brain Behav. 6, 389-400.   DOI
183 Kano, M., Ohno-Shosaku, T., Hashimotodani, Y., Uchigashima, M. and Watanabe, M. (2009) Endocannabinoid-mediated control of synaptic transmission. Physiol. Rev. 89, 309-380.   DOI
184 Karvat, G. and Kimchi, T. (2014) Acetylcholine elevation relieves cognitive rigidity and social deficiency in a mouse model of autism. Neuropsychopharmacology 39, 831-840.   DOI
185 Savaskan, E., Ehrhardt, R., Schulz, A., Walter, M. and Schachinger, H. (2008) Post-learning intranasal oxytocin modulates human memory for facial identity. Psychoneuroendocrinology 33, 368-374.   DOI
186 Scattoni, M. L., McFarlane, H. G., Zhodzishsky, V., Caldwell, H. K., Young, W. S., Ricceri, L. and Crawley, J. N. (2008) Reduced ultrasonic vocalizations in vasopressin 1b knockout mice. Behav. Brain Res. 187, 371-378.   DOI
187 Schaaf, C. P., Gonzalez-Garay, M. L., Xia, F., Potocki, L., Gripp, K. W., Zhang, B., Peters, B. A., McElwain, M. A., Drmanac, R., Beaudet, A. L., Caskey, C. T. and Yang, Y. (2013) Truncating mutations of MAGEL2 cause Prader-Willi phenotypes and autism. Nat. Genet. 45, 1405-1408.   DOI
188 Schaller, F., Watrin, F., Sturny, R., Massacrier, A., Szepetowski, P. and Muscatelli, F. (2010) A single postnatal injection of oxytocin rescues the lethal feeding behaviour in mouse newborns deficient for the imprinted Magel2 gene. Hum. Mol. Genet. 19, 4895-4905.   DOI
189 Schmale, H., Borowiak, B., Holtgreve-Grez, H. and Richter, D. (1989) Impact of altered protein structures on the intracellular traffic of a mutated vasopressin precursor from Brattleboro rats. Eur. J. Biochem. 182, 621-627.   DOI
190 Schmeisser, M. J., Ey, E., Wegener, S., Bockmann, J., Stempel, A. V., Kuebler, A., Janssen, A.-L., Udvardi, P. T., Shiban, E., Spilker, C., Balschun, D., Skryabin, B. V., Dieck, S. t., Smalla, K. H., Montag, D., Leblond, C. S., Faure, P., Torquet, N., Le Sourd, A. M., Toro, R., Grabrucker, A. M., Shoichet, S. A., Schmitz, D., Kreutz, M. R., Bourgeron, T., Gundelfinger, E. D. and Boeckers, T. M. (2012) Autistic-like behaviours and hyperactivity in mice lacking ProSAP1/ Shank2. Nature 486, 256-260.   DOI
191 Carson, R. P., Van Nielen, D. L., Winzenburger, P. A. and Ess, K. C. (2012) Neuronal and glia abnormalities in Tsc1-deficient forebrain and partial rescue by rapamycin. Neurobiol. Dis. 45, 369-380.   DOI
192 Casanova, M. F., Buxhoeveden, D. and Gomez, J. (2003) Disruption in the inhibitory architecture of the cell minicolumn: implications for autisim. Neuroscientist 9, 496-507.   DOI
193 Casanova, M. F., Buxhoeveden, D. P. and Brown, C. (2002) Clinical and macroscopic correlates of minicolumnar pathology in autism. J. Child Neurol. 17, 692-695.   DOI
194 Casanova, M. F., van Kooten, I. A., Switala, A. E., van Engeland, H., Heinsen, H., Steinbusch, H. W., Hof, P. R., Trippe, J., Stone, J. and Schmitz, C. (2006) Minicolumnar abnormalities in autism. Acta Neuropathol. 112, 287-303.   DOI
195 Cases, O., Seif, I., Grimsby, J., Gaspar, P., Chen, K., Pournin, S., Muller, U., Aguet, M., Babinet, C., Shih, J. C. and De Maeyer, E. (1995) Aggressive behavior and altered amounts of brain serotonin and norepinephrine in mice lacking MAOA. Science 268, 1763-1766.   DOI
196 Chadman, K. K. (2011) Fluoxetine but not risperidone increases sociability in the BTBR mouse model of autism. Pharmacol. Biochem. Behav. 97, 586-594.   DOI
197 Chahrour, M., Jung, S. Y., Shaw, C., Zhou, X., Wong, S. T., Qin, J. and Zoghbi, H. Y. (2008) MeCP2, a key contributor to neurological disease, activates and represses transcription. Science 320, 1224-1229.   DOI
198 Chahrour, M. and Zoghbi, H. Y. (2007) The story of Rett syndrome: from clinic to neurobiology. Neuron 56, 422-437.   DOI
199 Chakrabarti, S. and Fombonne, E. (2005) Pervasive developmental disorders in preschool children: confirmation of high prevalence. Am. J. Psychiatry 162, 1133-1141.   DOI
200 Kataoka, S., Takuma, K., Hara, Y., Maeda, Y., Ago, Y. and Matsuda, T. (2013) Autism-like behaviours with transient histone hyperacetylation in mice treated prenatally with valproic acid. Int. J. Neuropsychopharmacol. 16, 91-103.   DOI
201 Kavaliers, M., Colwell, D. D., Choleris, E., Agmo, A., Muglia, L. J., Ogawa, S. and Pfaff, D. W. (2003) Impaired discrimination of and aversion to parasitized male odors by female oxytocin knockout mice. Genes Brain Behav. 2, 220-230.   DOI
202 Kemper, T. L. and Bauman, M. (1998) Neuropathology of infantile autism. J. Neuropathol. Exp. Neurol. 57, 645-652.   DOI
203 Kemper, T. L. and Bauman, M. L. (1993) The contribution of neuropathologic studies to the understanding of autism. Neurol. Clin. 11, 175-187.   DOI
204 Kim, H. J. and Thayer, S. A. (2009) Lithium increases synapse formation between hippocampal neurons by depleting phosphoinositides. Mol. Pharmacol. 75, 1021-1030.   DOI
205 Kim, J. W., Seung, H., Kwon, K. J., Ko, M. J., Lee, E. J., Oh, H. A., Choi, C. S., Kim, K. C., Gonzales, E. L., You, J. S., Choi, D. H., Lee, J., Han, S. H., Yang, S. M., Cheong, J. H., Shin, C. Y. and Bahn, G. H. (2014a) Subchronic treatment of donepezil rescues impaired social, hyperactive, and stereotypic behavior in valproic acid-induced animal model of autism. PLoS One 9, e104927.   DOI
206 Kim, K. C., Choi, C. S., Kim, J.-W., Han, S.-H., Cheong, J. H., Ryu, J. H. and Shin, C. Y. (2014b) MeCP2 Modulates Sex Differences in the Postsynaptic Development of the Valproate Animal Model of Autism. Mol. Neurobiol. 1-17.
207 Kim, K. C., Kim, P., Go, H. S., Choi, C. S., Yang, S. I., Cheong, J. H., Shin, C. Y. and Ko, K. H. (2011) The critical period of valproate exposure to induce autistic symptoms in Sprague-Dawley rats. Toxicol. Lett. 201, 137-142.   DOI
208 Schneider, M. and Koch, M. (2005) Deficient social and play behavior in juvenile and adult rats after neonatal cortical lesion: effects of chronic pubertal cannabinoid treatment. Neuropsychopharmacology 30, 944-957.   DOI
209 Schneider, T., Roman, A., Basta-Kaim, A., Kubera, M., Budziszewska, B., Schneider, K. and Przewlocki, R. (2008) Gender-specific behavioral and immunological alterations in an animal model of autism induced by prenatal exposure to valproic acid. Psychoneuroendocrinology 33, 728-740.   DOI
210 Schreck, K. A., Mulick, J. A. and Smith, A. F. (2004) Sleep problems as possible predictors of intensified symptoms of autism. Res. Dev. Disabil. 25, 57-66.   DOI
211 Schumann, C. M., Hamstra, J., Goodlin-Jones, B. L., Lotspeich, L. J., Kwon, H., Buonocore, M. H., Lammers, C. R., Reiss, A. L. and Amaral, D. G. (2004) The amygdala is enlarged in children but not adolescents with autism; the hippocampus is enlarged at all ages. J. Neurosci. 24, 6392-6401.   DOI
212 Scott-Van Zeeland, A. A., Abrahams, B. S., Alvarez-Retuerto, A. I., Sonnenblick, L. I., Rudie, J. D., Ghahremani, D., Mumford, J. A., Poldrack, R. A., Dapretto, M., Geschwind, D. H. and Bookheimer, S. Y. (2010) Altered functional connectivity in frontal lobe circuits is associated with variation in the autism risk gene CNTNAP2. Sci. Transl. Med. 2, 56ra80.
213 Shinohe, A., Hashimoto, K., Nakamura, K., Tsujii, M., Iwata, Y., Tsuchiya, K. J., Sekine, Y., Suda, S., Suzuki, K., Sugihara, G., Matsuzaki, H., Minabe, Y., Sugiyama, T., Kawai, M., Iyo, M., Takei, N. and Mori, N. (2006) Increased serum levels of glutamate in adult patients with autism. Prog. Neuropsychopharmacol. Biol. Psychiatry 30, 1472-1477.   DOI
214 Silva, A. J. and Ehninger, D. (2009) Adult reversal of cognitive phenotypes in neurodevelopmental disorders. J. Neurodev. Disord. 1, 150-157.   DOI
215 Chan, J. P., Unger, T. J., Byrnes, J. and Rios, M. (2006) Examination of behavioral deficits triggered by targeting Bdnf in fetal or postnatal brains of mice. Neuroscience 142, 49-58.   DOI
216 Chao, H. T., Chen, H., Samaco, R. C., Xue, M., Chahrour, M., Yoo, J., Neul, J. L., Gong, S., Lu, H. C., Heintz, N., Ekker, M., Rubenstein, J. L., Noebels, J. L., Rosenmund, C. and Zoghbi, H. Y. (2010) Dysfunction in GABA signalling mediates autism-like stereotypies and Rett syndrome phenotypes. Nature 468, 263-269.   DOI
217 Cheh, M. A., Millonig, J. H., Roselli, L. M., Ming, X., Jacobsen, E., Kamdar, S. and Wagner, G. C. (2006) En2 knockout mice display neurobehavioral and neurochemical alterations relevant to autism spectrum disorder. Brain Res. 1116, 166-176.   DOI
218 Chen, Y., Beffert, U., Ertunc, M., Tang, T. S., Kavalali, E. T., Bezprozvanny, I. and Herz, J. (2005) Reelin modulates NMDA receptor activity in cortical neurons. J. Neurosci. 25, 8209-8216.   DOI
219 Chess, S., Fernandez, P. and Korn, S. (1978) Behavioral consequences of congenital rubella. J. Pediatr. 93, 699-703.   DOI
220 Chevere-Torres, I., Maki, J. M., Santini, E. and Klann, E. (2012) Impaired social interactions and motor learning skills in tuberous sclerosis complex model mice expressing a dominant/negative form of tuberin. Neurobiol. Dis. 45, 156-164.   DOI
221 Kim, K. C., Lee, D.-K., Go, H. S., Kim, P., Choi, C. S., Kim, J.-W., Jeon, S. J., Song, M.-R. and Shin, C. Y. (2014c) Pax6-dependent cortical glutamatergic neuronal differentiation regulates autism-like behavior in prenatally valproic acid-exposed rat offspring. Mol. Neurobiol. 49, 512-528.   DOI
222 Kolevzon, A., Cai, G., Soorya, L., Takahashi, N., Grodberg, D., Kajiwara, Y., Willner, J. P., Tryfon, A. and Buxbaum, J. D. (2011) Analysis of a purported SHANK3 mutation in a boy with autism: Clinical impact of rare variant research in neurodevelopmental disabilities. Brain Res. 1380, 98-105.   DOI
223 Kolevzon, A., Gross, R. and Reichenberg, A. (2007) Prenatal and perinatal risk factors for autism: a review and integration of findings. Arch. Pediatr. Adolesc. Med. 161, 326-333.   DOI
224 Korade, Z., Folkes, O. M. and Harrison, F. E. (2013) Behavioral and serotonergic response changes in the Dhcr7-HET mouse model of Smith-Lemli-Opitz syndrome. Pharmacol. Biochem. Behav. 106, 101-108.   DOI
225 Kuemerle, B., Gulden, F., Cherosky, N., Williams, E. and Herrup, K. (2007) The mouse Engrailed genes: a window into autism. Behav. Brain Res. 176, 121-132.   DOI
226 Kuemerle, B., Zanjani, H., Joyner, A. and Herrup, K. (1997) Pattern deformities and cell loss in Engrailed-2 mutant mice suggest two separate patterning events during cerebellar development. J. Neurosci. 17, 7881-7889.   DOI
227 Kumamaru, E., Egashira, Y., Takenaka, R. and Takamori, S. (2014) Valproic acid selectively suppresses the formation of inhibitory synapses in cultured cortical neurons. Neurosci. Lett. 569, 142-147.   DOI
228 Kuwagata, M., Ogawa, T., Shioda, S. and Nagata, T. (2009) Observation of fetal brain in a rat valproate-induced autism model: a developmental neurotoxicity study. Int. J. Dev. Neurosci. 27, 399-405.   DOI
229 Silva, A. J., Frankland, P. W., Marowitz, Z., Friedman, E., Laszlo, G. S., Cioffi, D., Jacks, T. and Bourtchuladze, R. (1997) A mouse model for the learning and memory deficits associated with neurofibromatosis type I. Nat. Genet. 15, 281-284.   DOI
230 Silverman, J. L., Tolu, S. S., Barkan, C. L. and Crawley, J. N. (2010) Repetitive self-grooming behavior in the BTBR mouse model of autism is blocked by the mGluR5 antagonist MPEP. Neuropsychopharmacology 35, 976-989.   DOI
231 Silverman, J. L., Turner, S. M., Barkan, C. L., Tolu, S. S., Saxena, R., Hung, A. Y., Sheng, M. and Crawley, J. N. (2011) Sociability and motor functions in Shank1 mutant mice. Brain Res. 1380, 120-137.   DOI
232 Singh, C., Bortolato, M., Bali, N., Godar, S. C., Scott, A. L., Chen, K., Thompson, R. F. and Shih, J. C. (2013) Cognitive abnormalities and hippocampal alterations in monoamine oxidase A and B knockout mice. Proc. Natl. Acad. Sci. U.S.A. 110, 12816-12821.   DOI
233 Singh, N. N., Lancioni, G. E., Winton, A. S., Fisher, B. C., Wahler, R. G., Mcaleavey, K., Singh, J. and Sabaawi, M. (2006) Mindful parenting decreases aggression, noncompliance, and self-injury in children with autism. J. Emot. Behav. Disord. 14, 169-177.   DOI
234 Sinkkonen, S. T., Homanics, G. E. and Korpi, E. R. (2003) Mouse models of Angelman syndrome, a neurodevelopmental disorder, display different brain regional GABA(A) receptor alterations. Neurosci. Lett. 340, 205-208.   DOI
235 Skidmore, B. J., Chiller, J. M., Morrison, D. C. and Weigle, W. O. (1975) Immunologic properties of bacterial lipopolysaccharide (LPS): correlation between the mitogenic, adjuvant, and immunogenic activities. J. Immunol. 114, 770-775.
236 Ching, M. S., Shen, Y., Tan, W. H., Jeste, S. S., Morrow, E. M., Chen, X., Mukaddes, N. M., Yoo, S. Y., Hanson, E., Hundley, R., Austin, C., Becker, R. E., Berry, G. T., Driscoll, K., Engle, E. C., Friedman, S., Gusella, J. F., Hisama, F. M., Irons, M. B., Lafiosca, T., LeClair, E., Miller, D. T., Neessen, M., Picker, J. D., Rappaport, L., Rooney, C. M., Sarco, D. P., Stoler, J. M., Walsh, C. A., Wolff, R. R., Zhang, T., Nasir, R. H. and Wu, B. L. (2010) Deletions of NRXN1 (neurexin- 1) predispose to a wide spectrum of developmental disorders. Am. J. Med. Genet. B Neuropsychiatr. Genet. 153B, 937-947.
237 Cho, K. S., Kwon, K. J., Choi, C. S., Jeon, S. J., Kim, K. C., Park, J. H., Ko, H. M., Lee, S. H., Cheong, J. H., Ryu, J. H., Han, S. H. and Shin, C. Y. (2013) Valproic acid induces astrocyte-dependent neurite outgrowth from cultured rat primary cortical neuron via modulation of tPA/PAI-1 activity. Glia 61, 694-709.   DOI
238 Christianson, A. L., Chesler, N. and Kromberg, J. G. (1994) Fetal valproate syndrome: clinical and neuro-developmental features in two sibling pairs. Dev. Med. Child Neurol. 36, 361-369.
239 Chu, E. C. and Tarnawski, A. S. (2004) PTEN regulatory functions in tumor suppression and cell biology. Med. Sci. Monit. 10, RA235-RA241.
240 Cichowski, K. and Jacks, T. (2001) NF1 tumor suppressor gene function: narrowing the GAP. Cell 104, 593-604.   DOI
241 Cohen, I., Liu, X., Lewis, M., Chudley, A., Forster-Gibson, C., Gonzalez, M., Jenkins, E., Brown, W. and Holden, J. (2011) Autism severity is associated with child and maternal MAOA genotypes. Clin. Genet. 79, 355-362.   DOI
242 Cohen, I. L., Liu, X., Schutz, C., White, B. N., Jenkins, E. C., Brown, W. T. and Holden, J. J. (2003) Association of autism severity with a monoamine oxidase A functional polymorphism. Clin. Genet. 64, 190-197.   DOI
243 Kwon, C. H., Luikart, B. W., Powell, C. M., Zhou, J., Matheny, S. A., Zhang, W., Li, Y., Baker, S. J. and Parada, L. F. (2006) Pten regulates neuronal arborization and social interaction in mice. Neuron 50, 377-388.   DOI
244 Lante, F., Meunier, J., Guiramand, J., De Jesus Ferreira, M. C., Cambonie, G., Aimar, R., Cohen-Solal, C., Maurice, T., Vignes, M. and Barbanel, G. (2008) Late N-acetylcysteine treatment prevents the deficits induced in the offspring of dams exposed to an immune stress during gestation. Hippocampus 18, 602-609.   DOI
245 Larimore, J. L., Chapleau, C. A., Kudo, S., Theibert, A., Percy, A. K. and Pozzo-Miller, L. (2009) Bdnf overexpression in hippocampal neurons prevents dendritic atrophy caused by Rett-associated MECP2 mutations. Neurobiol. Dis. 34, 199-211.
246 Leblond, C. S., Heinrich, J., Delorme, R., Proepper, C., Betancur, C., Huguet, G., Konyukh, M., Chaste, P., Ey, E., Rastam, M., Anckars?ter, H., Nygren, G., Gillberg, I. C., Melke, J., Toro, R., Regnault, B., Fauchereau, F., Mercati, O., Lemi?re, N., Skuse, D., Poot, M., Holt, R., Monaco, A. P., Järvelä, I., Kantojärvi, K., Vanhala, R., Curran, S., Collier, D. A., Bolton, P., Chiocchetti, A., Klauck, S. M., Poustka, F., Freitag, C. M., Waltes, R., Kopp, M., Duketis, E., Bacchelli, E., Minopoli, F., Ruta, L., Battaglia, A., Mazzone, L., Maestrini, E., Sequeira, A. F., Oliveira, B., Vicente, A., Oliveira, G., Pinto, D., Scherer, S. W., Zelenika, D., Delepine, M., Lathrop, M., Bonneau, D., Guinchat, V., Devillard, F., Assouline, B., Mouren, M. C., Leboyer, M., Gillberg, C., Boeckers, T. M. and Bourgeron, T. (2012) Genetic and functional analyses of SHANK2 mutations suggest a multiple hit model of autism spectrum disorders. PLoS Genet. 8, e1002521.   DOI
247 Lee, B. K. and McGrath, J. J. (2015) Advancing parental age and autism: multifactorial pathways. Trends Mol. Med. 21, 118-125.   DOI
248 Smalheiser, N. R., Costa, E., Guidotti, A., Impagnatiello, F., Auta, J., Lacor, P., Kriho, V. and Pappas, G. D. (2000) Expression of reelin in adult mammalian blood, liver, pituitary pars intermedia, and adrenal chromaffin cells. Proc. Natl. Acad. Sci. U.S.A. 97, 1281-1286.   DOI
249 Smith, S. E., Li, J., Garbett, K., Mirnics, K. and Patterson, P. H. (2007) Maternal immune activation alters fetal brain development through interleukin-6. J. Neurosci. 27, 10695-10702.   DOI
250 Smith, S. E., Zhou, Y. D., Zhang, G., Jin, Z., Stoppel, D. C. and Anderson, M. P. (2011) Increased gene dosage of Ube3a results in autism traits and decreased glutamate synaptic transmission in mice. Sci. Transl. Med. 3, 103ra97.
251 Spence, S. J. and Schneider, M. T. (2009) The role of epilepsy and epileptiform EEGs in autism spectrum disorders. Pediatr. Res. 65, 599-606.   DOI
252 Spencer, C. M., Alekseyenko, O., Serysheva, E., Yuva-Paylor, L. A. and Paylor, R. (2005) Altered anxiety-related and social behaviors in the Fmr1 knockout mouse model of fragile X syndrome. Genes Brain Behav. 4, 420-430.   DOI
253 Stanfield, B. B. and Cowan, W. M. (1979) The morphology of the hippocampus and dentate gyrus in normal and reeler mice. J. Comp. Neurol. 185, 393-422.   DOI
254 Steffenburg, S., Gillberg, C., Hellgren, L., Andersson, L., Gillberg, I. C., Jakobsson, G. and Bohman, M. (1989) A twin study of autism in Denmark, Finland, Iceland, Norway and Sweden. J. Child Psychol. Psychiatry 30, 405-416.   DOI
255 Comoletti, D., De Jaco, A., Jennings, L. L., Flynn, R. E., Gaietta, G., Tsigelny, I., Ellisman, M. H. and Taylor, P. (2004) The Arg451Cysneuroligin-3 mutation associated with autism reveals a defect in protein processing. J. Neurosci. 24, 4889-4893.   DOI
256 Correa-Cerro, L. S., Wassif, C. A., Kratz, L., Miller, G. F., Munasinghe, J. P., Grinberg, A., Fliesler, S. J. and Porter, F. D. (2006) Development and characterization of a hypomorphic Smith-Lemli-Opitz syndrome mouse model and efficacy of simvastatin therapy. Hum. Mol. Genet. 15, 839-851.   DOI
257 Costa, R. M., Federov, N. B., Kogan, J. H., Murphy, G. G., Stern, J., Ohno, M., Kucherlapati, R., Jacks, T. and Silva, A. J. (2002) Mechanism for the learning deficits in a mouse model of neurofibromatosis type 1. Nature 415, 526-530.   DOI
258 Costa, R. M., Yang, T., Huynh, D. P., Pulst, S. M., Viskochil, D. H., Silva, A. J. and Brannan, C. I. (2001) Learning deficits, but normal development and tumor predisposition, in mice lacking exon 23a of Nf1. Nat. Genet. 27, 399-405.   DOI
259 Courchesne, E., Saitoh, O., Yeung-Courchesne, R., Press, G. A., Lincoln, A. J., Haas, R. H. and Schreibman, L. (1994) Abnormality of cerebellar vermian lobules VI and VII in patients with infantile autism: identification of hypoplastic and hyperplastic subgroups with MR imaging. AJR Am. J. Roentgenol. 162, 123-130.   DOI
260 Croen, L. A., Grether, J. K., Yoshida, C. K., Odouli, R. and Van de Water, J. (2005) Maternal autoimmune diseases, asthma and allergies, and childhood autism spectrum disorders: a case-control study. Arch. Pediatr. Adolesc. Med. 159, 151-157.
261 Curatolo, P., Porfirio, M. C., Manzi, B. and Seri, S. (2004) Autism in tuberous sclerosis. Eur. J. Paediatr. Neurol. 8, 327-332.   DOI
262 Lee, H. J., Caldwell, H. K., Macbeth, A. H. and Young, W. S., 3rd (2008) Behavioural studies using temporal and spatial inactivation of the oxytocin receptor. Prog. Brain Res. 170, 73-77.   DOI
263 Leyfer, O. T., Folstein, S. E., Bacalman, S., Davis, N. O., Dinh, E., Morgan, J., Tager-Flusberg, H. and Lainhart, J. E. (2006) Comorbid psychiatric disorders in children with autism: Interview development and rates of disorders. J. Autism Dev. Disord. 36, 849-861.   DOI
264 Li, X., Hu, Z., He, Y., Xiong, Z., Long, Z., Peng, Y., Bu, F., Ling, J., Xun, G. and Mo, X. (2010) Association analysis of CNTNAP2 polymorphisms with autism in the Chinese Han population. Psychiatr. Genet. 20, 113-117.
265 Li, X., Zhang, J., Cao, Z., Wu, J. and Shi, Y. (2006) Solution structure of GOPC PDZ domain and its interaction with the C-terminal motif of neuroligin. Protein Sci. 15, 2149-2158.   DOI
266 Liu, W. S., Pesold, C., Rodriguez, M. A., Carboni, G., Auta, J., Lacor, P., Larson, J., Condie, B. G., Guidotti, A. and Costa, E. (2001) Down-regulation of dendritic spine and glutamic acid decarboxylase 67 expressions in the reelin haploinsufficient heterozygous reeler mouse. Proc. Natl. Acad. Sci. U.S.A. 98, 3477-3482.   DOI
267 Liu, X., Kawamura, Y., Shimada, T., Otowa, T., Koishi, S., Sugiyama, T., Nishida, H., Hashimoto, O., Nakagami, R., Tochigi, M., Umekage, T., Kano, Y., Miyagawa, T., Kato, N., Tokunaga, K. and Sasaki, T. (2010) Association of the oxytocin receptor (OXTR) gene polymorphisms with autism spectrum disorder (ASD) in the Japanese population. J. Hum. Genet. 55, 137-141.   DOI
268 Liu, Z. H. and Smith, C. B. (2009) Dissociation of social and nonsocial anxiety in a mouse model of fragile X syndrome. Neurosci. Lett. 454, 62-66.   DOI
269 Stephenson, D. T., O'Neill, S. M., Narayan, S., Tiwari, A., Arnold, E., Samaroo, H. D., Du, F., Ring, R. H., Campbell, B., Pletcher, M., Vaidya, V. A. and Morton, D. (2011) Histopathologic characterization of the BTBR mouse model of autistic-like behavior reveals selective changes in neurodevelopmental proteins and adult hippocampal neurogenesis. Mol. Autism 2, 7.   DOI
270 Strang, J. F., Kenworthy, L., Daniolos, P., Case, L., Wills, M. C., Martin, A. and Wallace, G. L. (2012) Depression and Anxiety Symptoms in Children and Adolescents with Autism Spectrum Disorders without Intellectual Disability. Res. Autism Spectr. Disord. 6, 406-412.   DOI
271 Strauss, K. A., Puffenberger, E. G., Huentelman, M. J., Gottlieb, S., Dobrin, S. E., Parod, J. M., Stephan, D. A. and Morton, D. H. (2006) Recessive symptomatic focal epilepsy and mutant contactin-associated protein-like 2. N. Engl. J. Med. 354, 1370-1377.   DOI
272 Stromland, K., Philipson, E. and Andersson Gronlund, M. (2002) Offspring of male and female parents with thalidomide embryopathy: birth defects and functional anomalies. Teratology 66, 115-121.   DOI
273 Sudhof, T. C. (2008) Neuroligins and neurexins link synaptic function to cognitive disease. Nature 455, 903-911.   DOI
274 Szatmari, P., Maziade, M., Zwaigenbaum, L., Merette, C., Roy, M. A., Joober, R. and Palmour, R. (2007) Informative phenotypes for genetic studies of psychiatric disorders. Am. J. Med. Genet. B Neuropsychiatr. Genet. 144B, 581-588.   DOI
275 Taieb, O., Baleyte, J. M., Mazet, P. and Fillet, A. M. (2001) Borna disease virus and psychiatry. Eur. Psychiatry 16, 3-10.
276 D'Arcangelo, G. (2005) The reeler mouse: anatomy of a mutant. Int. Rev. Neurobiol. 71, 383-417.   DOI
277 Dani, V. S., Chang, Q., Maffei, A., Turrigiano, G. G., Jaenisch, R. and Nelson, S. B. (2005) Reduced cortical activity due to a shift in the balance between excitation and inhibition in a mouse model of Rett syndrome. Proc. Natl. Acad. Sci. U.S.A. 102, 12560-12565.   DOI
278 Daniels, J. L., Forssen, U., Hultman, C. M., Cnattingius, S., Savitz, D. A., Feychting, M. and Sparen, P. (2008) Parental psychiatric disorders associated with autism spectrum disorders in the offspring. Pediatrics 121, e1357-e1362.   DOI
279 Davis, L. K., Hazlett, H. C., Librant, A. L., Nopoulos, P., Sheffield, V. C., Piven, J. and Wassink, T. H. (2008) Cortical enlargement in autism is associated with a functional VNTR in the monoamine oxidase A gene. Am. J. Med. Genet. B Neuropsychiatr. Genet. 147B, 1145-1151.   DOI
280 Daws, L. C., Munn, J. L., Valdez, M. F., Frosto-Burke, T. and Hensler, J. G. (2007) Serotonin transporter function, but not expression, is dependent on brain-derived neurotrophic factor (BDNF): in vivo studies in BDNF-deficient mice. J. Neurochem. 101, 641-651.   DOI
281 DeLorey, T. M., Handforth, A., Anagnostaras, S. G., Homanics, G. E., Minassian, B. A., Asatourian, A., Fanselow, M. S., Delgado-Escueta, A., Ellison, G. D. and Olsen, R. W. (1998) Mice lacking the beta3 subunit of the GABAA receptor have the epilepsy phenotype and many of the behavioral characteristics of Angelman syndrome. J. Neurosci. 18, 8505-8514.   DOI
282 DeLorey, T. M., Sahbaie, P., Hashemi, E., Homanics, G. E. and Clark, J. D. (2008) Gabrb3 gene deficient mice exhibit impaired social and exploratory behaviors, deficits in non-selective attention and hypoplasia of cerebellar vermal lobules: a potential model of autism spectrum disorder. Behav. Brain Res. 187, 207-220.   DOI
283 LoParo, D. and Waldman, I. D. (2015) The oxytocin receptor gene (OXTR) is associated with autism spectrum disorder: a meta-analysis. Mol. Psychiatry 20, 640-646.   DOI
284 Lozovaya, N., Gataullina, S., Tsintsadze, T., Tsintsadze, V., Pallesi-Pocachard, E., Minlebaev M., Goriounova, N. A., Buhler, E., Watrin, F., Shityakov, S., Becker, A. J., Bordey, A., Milh, M., Scavarda, D., Bulteau, C., Dorfmuller, G., Delalande, O., Represa, A., Cardoso, C., Dulac, O., Ben-Ari, Y. and Burnashev, N. (2014) Selective suppression of excessive GluN2C expression rescues early epilepsy in a tuberous sclerosis murine model. Nat. Commun. 5, 4563.   DOI
285 Luikart, B. W., Schnell, E., Washburn, E. K., Bensen, A. L., Tovar, K. R. and Westbrook, G. L. (2011) Pten knockdown in vivo increases excitatory drive onto dentate granule cells. J. Neurosci. 31, 4345-4354.   DOI
286 Lush, M. E., Li, Y., Kwon, C. H., Chen, J. and Parada, L. F. (2008) Neurofibromin is required for barrel formation in the mouse somatosensory cortex. J. Neurosci. 28, 1580-1587.   DOI
287 Maccarrone, M., Rossi, S., Bari, M., De Chiara, V., Rapino, C., Musella, A., Bernardi, G., Bagni, C. and Centonze, D. (2010) Abnormal mGlu 5 receptor/endocannabinoid coupling in mice lacking FMRP and BC1 RNA. Neuropsychopharmacology 35, 1500-1509.   DOI
288 MacQueen, G. M., Ramakrishnan, K., Croll, S. D., Siuciak, J. A., Yu, G., Young, L. T. and Fahnestock, M. (2001) Performance of heterozygous brain-derived neurotrophic factor knockout mice on behavioral analogues of anxiety, nociception, and depression. Behav. Neurosci. 115, 1145-1153.   DOI
289 Maezawa, I. and Jin, L. W. (2010) Rett syndrome microglia damage dendrites and synapses by the elevated release of glutamate. J. Neurosci. 30, 5346-5356.   DOI
290 Tang, G., Gudsnuk, K., Kuo, S.-H., Cotrina, M. L., Rosoklija, G., Sosunov, A., Sonders, M. S., Kanter, E., Castagna, C., Yamamoto, A., Yue, Z., Arancio, O., Peterson, B. S., Champagne, F., Dwork, A. J., Goldman, J. and Sulzer, D. (2014) Loss of mTOR-Dependent Macroautophagy Causes Autistic-like Synaptic Pruning Deficits. Neuron 83, 1131-1143.   DOI
291 Tansey, K. E., Brookes, K. J., Hill, M. J., Cochrane, L. E., Gill, M., Skuse, D., Correia, C., Vicente, A., Kent, L., Gallagher, L. and Anney, R. J. (2010) Oxytocin receptor (OXTR) does not play a major role in the aetiology of autism: genetic and molecular studies. Neurosci. Lett. 474, 163-167.   DOI
292 Taurines, R., Segura, M., Schecklmann, M., Albantakis, L., Grunblatt, E., Walitza, S., Jans, T., Lyttwin, B., Haberhausen, M., Theisen, F. M., Martin, B., Briegel, W., Thome, J., Schwenck, C., Romanos, M. and Gerlach, M. (2014) Altered peripheral BDNF mRNA expression and BDNF protein concentrations in blood of children and adolescents with autism spectrum disorder. J. Neural Transm. (Vienna) 121, 1117-1128.   DOI
293 Tavassoli, T., Auyeung, B., Murphy, L. C., Baron-Cohen, S. and Chakrabarti, B. (2012) Variation in the autism candidate gene GABRB3 modulates tactile sensitivity in typically developing children. Mol. Autism 3, 6.   DOI
294 The International Schizophrenia Consortium (2008) Rare chromosomal deletions and duplications increase risk of schizophrenia. Nature 455, 237-241.   DOI
295 Tierney, E., Bukelis, I., Thompson, R. E., Ahmed, K., Aneja, A., Kratz, L. and Kelley, R. I. (2006) Abnormalities of cholesterol metabolism in autism spectrum disorders. Am. J. Med. Genet. B Neuropsychiatr. Genet. 141B, 666-668.   DOI
296 Tierney, E., Nwokoro, N. A. and Kelley, R. I. (2000) Behavioral phenotype of RSH/Smith-Lemli-Opitz syndrome. Ment. Retard. Dev. Disabil. Res. Rev. 6, 131-134.   DOI
297 Di Cristofano, A., Pesce, B., Cordon-Cardo, C. and Pandolfi, P. P. (1998) Pten is essential for embryonic development and tumour suppression. Nat. Genet. 19, 348-355.   DOI
298 Diergaarde, L., Gerrits, M. A., Brouwers, J. P. and van Ree, J. M. (2005) Early amygdala damage disrupts performance on medial prefrontal cortex-related tasks but spares spatial learning and memory in the rat. Neuroscience 130, 581-590.   DOI
299 DiMario, F. J., Jr. (2004) Brain abnormalities in tuberous sclerosis complex. J. Child Neurol. 19, 650-657.   DOI
300 Dufour-Rainfray, D., Vourc'h, P., Le Guisquet, A. M., Garreau, L., Ternant, D., Bodard, S., Jaumain, E., Gulhan, Z., Belzung, C., Andres, C. R., Chalon, S. and Guilloteau, D. (2010) Behavior and serotonergic disorders in rats exposed prenatally to valproate: a model for autism. Neurosci. Lett. 470, 55-59.   DOI
301 Durand, C. M., Betancur, C., Boeckers, T. M., Bockmann, J., Chaste, P., Fauchereau, F., Nygren, G., Rastam, M., Gillberg, I. C., Anckarsater, H., Sponheim, E., Goubran-Botros, H., Delorme, R., Chabane, N., Mouren-Simeoni, M. C., de Mas, P., Bieth, E., Roge, B., Heron, D., Burglen, L., Gillberg, C., Leboyer, M. and Bourgeron, T. (2007) Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat. Genet. 39, 25-27.   DOI
302 Dykens, E. M., Lee, E. and Roof, E. (2011) Prader-Willi syndrome and autism spectrum disorders: an evolving story. J. Neurodev. Disord. 3, 225-237.   DOI
303 Ehninger, D., Sano, Y., de Vries, P. J., Dies, K., Franz, D., Geschwind, D. H., Kaur, M., Lee, Y. S., Li, W., Lowe, J. K., Nakagawa, J. A., Sahin, M., Smith, K., Whittemore, V. and Silva, A. J. (2012) Gestational immune activation and Tsc2 haploinsufficiency cooperate to disrupt fetal survival and may perturb social behavior in adult mice. Mol. Psychiatry 17, 62-70.   DOI
304 Manning, M. A., Cassidy, S. B., Clericuzio, C., Cherry, A. M., Schwartz, S., Hudgins, L., Enns, G. M. and Hoyme, H. E. (2004) Terminal 22q deletion syndrome: a newly recognized cause of speech and language disability in the autism spectrum. Pediatrics 114, 451-457.   DOI
305 Marrone, M. C., Marinelli, S., Biamonte, F., Keller, F., Sgobio, C. A., Ammassari-Teule, M., Bernardi, G. and Mercuri, N. B. (2006) Altered cortico-striatal synaptic plasticity and related behavioural impairments in reeler mice. Eur J. Neurosci. 24, 2061-2070.   DOI
306 Martin, H. G. and Manzoni, O. J. (2014) Late onset deficits in synaptic plasticity in the valproic acid rat model of autism. Front. Cell. Neurosci. 8, 23.
307 Martin, L. A., Escher, T., Goldowitz, D. and Mittleman, G. (2004) A relationship between cerebellar Purkinje cells and spatial working memory demonstrated in a lurcher/chimera mouse model system. Genes Brain Behav. 3, 158-166.   DOI
308 Martin, L. A., Goldowitz, D. and Mittleman, G. (2010) Repetitive behavior and increased activity in mice with Purkinje cell loss: a model for understanding the role of cerebellar pathology in autism. Eur. J. Neurosci. 31, 544-555.   DOI
309 Martin, M. R. (1981) Morphology of the cochlear nucleus of the normal and reeler mutant mouse. J. Comp. Neurol. 197, 141-152.   DOI
310 Martins, Y., Young, R. L. and Robson, D. C. (2008) Feeding and eating behaviors in children with autism and typically developing children. J. Autism Dev. Disord. 38, 1878-1887.   DOI
311 Marui, T., Hashimoto, O., Nanba, E., Kato, C., Tochigi, M., Umekage, T., Ishijima, M., Kohda, K., Kato, N. and Sasaki, T. (2004) Association between the neurofibromatosis-1 (NF1) locus and autism in the Japanese population. Am. J. Med. Genet. B Neuropsychiatr. Genet. 131B, 43-47.   DOI
312 Tottenham, N., Hertzig, M. E., Gillespie-Lynch, K., Gilhooly, T., Millner, A. J. and Casey, B. (2014) Elevated amygdala response to faces and gaze aversion in autism spectrum disorder. Soc. Cogn. Affect. Neurosci. 9, 106-117.   DOI
313 Truong, D. T., Rendall, A. R., Castelluccio, B. C., Eigsti, I. M. and Fitch, R. H. (2015) Auditory processing and morphological anomalies in medial geniculate nucleus of Cntnap2 mutant mice. Behav. Neurosci. 129, 731-743.   DOI
314 Tsai, P. T., Chu, Y., Greene-Colozzi, E., Sadowski, A. R., Leech, J. M., Steinberg, J., Crawley, J. N., Regehr, W. G. and Sahin, M. (2012) Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant mice. Nature 488, 647-651.   DOI
315 Tsai, S. J. (2005) Is autism caused by early hyperactivity of brain-derived neurotrophic factor? Med. Hypotheses 65, 79-82.   DOI
316 Tueting, P., Costa, E., Dwivedi, Y., Guidotti, A., Impagnatiello, F., Manev, R. and Pesold, C. (1999) The phenotypic characteristics of heterozygous reeler mouse. Neuroreport 10, 1329-1334.   DOI
317 Turrigiano, G. (2012) Homeostatic synaptic plasticity: local and global mechanisms for stabilizing neuronal function. Cold Spring Harb. Perspect. Biol. 4, a005736.
318 Tyzio, R., Nardou, R., Ferrari, D. C., Tsintsadze, T., Shahrokhi, A., Eftekhari, S., Khalilov, I., Tsintsadze, V., Brouchoud, C., Chazal, G., Lemonnier, E., Lozovaya, N., Burnashev, N. and Ben-Ari, Y. (2014) Oxytocin-mediated GABA inhibition during delivery attenuates autism pathogenesis in rodent offspring. Science 343, 675-679.   DOI
319 Uhlmann, E. J., Wong, M., Baldwin, R. L., Bajenaru, M. L., Onda, H., Kwiatkowski, D. J., Yamada, K. and Gutmann, D. H. (2002) Astrocyte- specific TSC1 conditional knockout mice exhibit abnormal neuronal organization and seizures. Ann. Neurol. 52, 285-296.   DOI
320 Engelmann, M. and Landgraf, R. (1994) Microdialysis administration of vasopressin into the septum improves social recognition in Brattleboro rats. Physiol. Behav. 55, 145-149.   DOI
321 Eslinger, P. J., Flaherty-Craig, C. V. and Benton, A. L. (2004) Developmental outcomes after early prefrontal cortex damage. Brain Cogn. 55, 84-103.   DOI
322 Etherton, M., Foldy, C., Sharma, M., Tabuchi, K., Liu, X., Shamloo, M., Malenka, R. C. and Sudhof, T. C. (2011) Autism-linked neuroligin- 3 R451C mutation differentially alters hippocampal and cortical synaptic function. Proc. Natl. Acad. Sci. U.S.A. 108, 13764-13769.   DOI
323 Etherton, M. R., Blaiss, C. A., Powell, C. M. and Sudhof, T. C. (2009) Mouse neurexin-1alpha deletion causes correlated electrophysiological and behavioral changes consistent with cognitive impairments. Proc. Natl. Acad. Sci. U.S.A. 106, 17998-18003.   DOI
324 Fatemi, S. H., Aldinger, K. A., Ashwood, P., Bauman, M. L., Blaha, C. D., Blatt, G. J., Chauhan, A., Chauhan, V., Dager, S. R., Dickson, P. E., Estes, A. M., Goldowitz, D., Heck, D. H., Kemper, T. L., King, B. H., Martin, L. A., Millen, K. J., Mittleman, G., Mosconi, M. W., Persico, A. M., Sweeney, J. A., Webb, S. J. and Welsh, J. P. (2012) Consensus paper: pathological role of the cerebellum in autism. Cerebellum 11, 777-807.   DOI
325 Fatemi, S. H., Halt, A. R., Stary, J. M., Kanodia, R., Schulz, S. C. and Realmuto, G. R. (2002) Glutamic acid decarboxylase 65 and 67 kDa proteins are reduced in autistic parietal and cerebellar cortices. Biol. Psychiatry 52, 805-810.   DOI
326 Fatemi, S. H., Reutiman, T. J., Folsom, T. D. and Thuras, P. D. (2009) GABA(A) receptor downregulation in brains of subjects with autism. J. Autism Dev. Disord. 39, 223-230.   DOI
327 Mazina, V., Gerdts, J., Trinh, S., Ankenman, K., Ward, T., Dennis, M. Y., Girirajan, S., Eichler, E. E. and Bernier, R. (2015) Epigenetics of autism-related impairment: copy number variation and maternal infection. J. Dev. Behav. Pediatr. 36, 61-67.   DOI
328 Mbarek, O., Marouillat, S., Martineau, J., Barthelemy, C., Muh, J. P. and Andres, C. (1999) Association study of the NF1 gene and autistic disorder. Am. J. Med. Genet. 88, 729-732.   DOI
329 McFarlane, H. G., Kusek, G. K., Yang, M., Phoenix, J. L., Bolivar, V. J. and Crawley, J. N. (2008) Autism-like behavioral phenotypes in BTBR T+tf/J mice. Genes Brain Behav. 7, 152-163.   DOI
330 McNaughton, C. H., Moon, J., Strawderman, M. S., Maclean, K. N., Evans, J. and Strupp, B. J. (2008) Evidence for social anxiety and impaired social cognition in a mouse model of fragile X syndrome. Behav. Neurosci. 122, 293-300.   DOI
331 Meikle, L., Talos, D. M., Onda, H., Pollizzi, K., Rotenberg, A., Sahin, M., Jensen, F. E. and Kwiatkowski, D. J. (2007) A mouse model of tuberous sclerosis: neuronal loss of Tsc1 causes dysplastic and ectopic neurons, reduced myelination, seizure activity, and limited survival. J. Neurosci. 27, 5546-5558.   DOI
332 Meyer, U., Feldon, J. and Dammann, O. (2011) Schizophrenia and autism: both shared and disorder-specific pathogenesis via perinatal inflammation? Pediatr. Res. 69, 26R-33R.   DOI
333 Meziane, H., Schaller, F., Bauer, S., Villard, C., Matarazzo, V., Riet, F., Guillon, G., Lafitte, D., Desarmenien, M. G., Tauber, M. and Muscatelli, F. (2015) An Early Postnatal Oxytocin Treatment Prevents Social and Learning Deficits in Adult Mice Deficient for Magel2, a Gene Involved in Prader-Willi Syndrome and Autism. Biol. Psychiatry 78, 85-94.   DOI
334 Veenstra-Vanderweele, J., Jessen, T. N., Thompson, B. J., Carter, M., Prasad, H. C., Steiner, J. A., Sutcliffe, J. S. and Blakely, R. D. (2009) Modeling rare gene variation to gain insight into the oldest biomarker in autism: construction of the serotonin transporter Gly56Ala knock-in mouse. J. Neurodev. Disord. 1, 158-171.   DOI
335 Verheij, C., Bakker, C. E., de Graaff, E., Keulemans, J., Willemsen, R., Verkerk, A. J., Galjaard, H., Reuser, A. J., Hoogeveen, A. T. and Oostra, B. A. (1993) Characterization and localization of the FMR-1 gene product associated with fragile X syndrome. Nature 363, 722-724.   DOI
336 Verpelli, C., Dvoretskova, E., Vicidomini, C., Rossi, F., Chiappalone, M., Schoen, M., Di Stefano, B., Mantegazza, R., Broccoli, V., Bockers, T. M., Dityatev, A. and Sala, C. (2011) Importance of Shank3 protein in regulating metabotropic glutamate receptor 5 (mGluR5) expression and signaling at synapses. J. Biol. Chem. 286, 34839-34850.   DOI
337 Volkmar, F. R. and Nelson, D. S. (1990) Seizure Disorders in Autism. J. Am. Acad. Child Adolesc. Psychiatry 29, 127-129.   DOI
338 Waage-Baudet, H., Lauder, J. M., Dehart, D. B., Kluckman, K., Hiller, S., Tint, G. S. and Sulik, K. K. (2003) Abnormal serotonergic development in a mouse model for the Smith-Lemli-Opitz syndrome: implications for autism. Int. J. Dev. Neurosci. 21, 451-459.   DOI
339 Wagstaff, J., Knoll, J. H., Fleming, J., Kirkness, E. F., Martin-Gallardo, A., Greenberg, F., Graham, J. M., Jr., Menninger, J., Ward, D., Venter, J. C. and Lalande, M. (1991) Localization of the gene encoding the GABAA receptor beta 3 subunit to the Angelman/Prader-Willi region of human chromosome 15. Am. J. Hum. Genet. 49, 330-337.
340 Ferguson, J. N., Aldag, J. M., Insel, T. R. and Young, L. J. (2001) Oxytocin in the medial amygdala is essential for social recognition in the mouse. J. Neurosci. 21, 8278-8285.   DOI
341 Fidler, D. J., Bailey, J. N. and Smalley, S. L. (2000) Macrocephaly in autism and other pervasive developmental disorders. Dev. Med. Child Neurol. 42, 737-740.   DOI
342 Fisher, S. E. and Scharff, C. (2009) FOXP2 as a molecular window into speech and language. Trends Genet. 25, 166-177.   DOI
343 Fitzky, B. U., Moebius, F. F., Asaoka, H., Waage-Baudet, H., Xu, L., Xu, G., Maeda, N., Kluckman, K., Hiller, S., Yu, H., Batta, A. K., Shefer, S., Chen, T., Salen, G., Sulik, K., Simoni, R. D., Ness, G. C., Glossmann, H., Patel, S. B. and Tint, G. S. (2001) 7-Dehydrocholesterol- dependent proteolysis of HMG-CoA reductase suppresses sterol biosynthesis in a mouse model of Smith-Lemli-Opitz/RSH syndrome. J. Clin. Invest. 108, 905-915.   DOI
344 Fombonne, E. (1999) The epidemiology of autism: a review. Psychol. Med. 29, 769-786.   DOI
345 Frye, C. A. and Llaneza, D. C. (2010) Corticosteroid and neurosteroid dysregulation in an animal model of autism, BTBR mice. Physiol. Behav. 100, 264-267.   DOI
346 Fukuchi, M., Nii, T., Ishimaru, N., Minamino, A., Hara, D., Takasaki, I., Tabuchi, A. and Tsuda, M. (2009) Valproic acid induces up- or down-regulation of gene expression responsible for the neuronal excitation and inhibition in rat cortical neurons through its epigenetic actions. Neurosci. Res. 65, 35-43.   DOI
347 Garber, K. B., Visootsak, J. and Warren, S. T. (2008) Fragile X syndrome. Eur. J. Hum. Genet. 16, 666-672.   DOI
348 Gardiner, S. M. and Bennett, T. (1983) The cardiovascular and renal responses to short-term isolation in Brattleboro rats. Clin. Sci. 64, 377-382.   DOI
349 Mineur, Y. S., Huynh, L. X. and Crusio, W. E. (2006) Social behavior deficits in the Fmr1 mutant mouse. Behav. Brain Res. 168, 172-175.   DOI
350 Miura, K., Kishino, T., Li, E., Webber, H., Dikkes, P., Holmes, G. L. and Wagstaff, J. (2002) Neurobehavioral and electroencephalographic abnormalities in Ube3a maternal-deficient mice. Neurobiol. Dis. 9, 149-159.   DOI
351 Monteggia, L. M., Luikart, B., Barrot, M., Theobold, D., Malkovska, I., Nef, S., Parada, L. F. and Nestler, E. J. (2007) Brain-derived neurotrophic factor conditional knockouts show gender differences in depression-related behaviors. Biol. Psychiatry 61, 187-197.   DOI
352 Moore, S. J., Turnpenny, P., Quinn, A., Glover, S., Lloyd, D. J., Montgomery, T. and Dean, J. C. (2000) A clinical study of 57 children with fetal anticonvulsant syndromes. J. Med. Genet. 37, 489-497.   DOI
353 Moretti, P., Levenson, J. M., Battaglia, F., Atkinson, R., Teague, R., Antalffy, B., Armstrong, D., Arancio, O., Sweatt, J. D. and Zoghbi, H. Y. (2006) Learning and memory and synaptic plasticity are impaired in a mouse model of Rett syndrome. J. Neurosci. 26, 319-327.   DOI
354 Moy, S. S., Nadler, J. J., Young, N. B., Nonneman, R. J., Grossman, A. W., Murphy, D. L., D'Ercole, A. J., Crawley, J. N., Magnuson, T. R. and Lauder, J. M. (2009) Social approach in genetically engineered mouse lines relevant to autism. Genes Brain Behav. 8, 129-142.   DOI
355 Murer, M. G., Yan, Q. and Raisman-Vozari, R. (2001) Brain-derived neurotrophic factor in the control human brain, and in Alzheimer's disease and Parkinson's disease. Prog. Neurobiol. 63, 71-124.   DOI
356 Wahlsten, D., Metten, P. and Crabbe, J. C. (2003) Survey of 21 inbred mouse strains in two laboratories reveals that BTBR T/+ tf/tf has severely reduced hippocampal commissure and absent corpus callosum. Brain Res. 971, 47-54.   DOI
357 Walcott, E. C., Higgins, E. A. and Desai, N. S. (2011) Synaptic and intrinsic balancing during postnatal development in rat pups exposed to valproic acid in utero. J. Neurosci. 31, 13097-13109.   DOI
358 Walsh, T., McClellan, J. M., McCarthy, S. E., Addington, A. M., Pierce, S. B., Cooper, G. M., Nord, A. S., Kusenda, M., Malhotra, D., Bhandari, A., Stray, S. M., Rippey, C. F., Roccanova, P., Makarov, V., Lakshmi, B., Findling, R. L., Sikich, L., Stromberg, T., Merriman, B., Gogtay, N., Butler, P., Eckstrand, K., Noory, L., Gochman, P., Long, R., Chen, Z., Davis, S., Baker, C., Eichler, E. E., Meltzer, P. S., Nelson, S. F., Singleton, A. B., Lee, M. K., Rapoport, J. L., King, M. C. and Sebat, J. (2008) Rare structural variants disrupt multiple genes in neurodevelopmental pathways in schizophrenia. Science 320, 539-543.   DOI
359 Wang, H., Meng, X. H., Ning, H., Zhao, X. F., Wang, Q., Liu, P., Zhang, H., Zhang, C., Chen, G. H. and Xu, D. X. (2010) Age- and genderdependent impairments of neurobehaviors in mice whose mothers were exposed to lipopolysaccharide during pregnancy. Toxicol. Lett. 192, 245-251.   DOI
360 Wang, S. S., Kloth, A. D. and Badura, A. (2014) The Cerebellum, Sensitive Periods, and Autism. Neuron 83, 518-532.   DOI
361 Warrier, V., Baron-Cohen, S. and Chakrabarti, B. (2013) Genetic variation in GABRB3 is associated with Asperger syndrome and multiple endophenotypes relevant to autism. Mol. Autism 4, 48.   DOI
362 Wassink, T. H., Piven, J., Vieland, V. J., Pietila, J., Goedken, R. J., Folstein, S. E. and Sheffield, V. C. (2004) Examination of AVPR1a as an autism susceptibility gene. Mol. Psychiatry 9, 968-972.   DOI