Skip to main content
Top
Published in: Molecular Autism 1/2016

Open Access 01-12-2016 | Research

Overexpression of Homer1a in the basal and lateral amygdala impairs fear conditioning and induces an autism-like social impairment

Authors: Anwesha Banerjee, Jonathan A. Luong, Anthony Ho, Aeshah O. Saib, Jonathan E. Ploski

Published in: Molecular Autism | Issue 1/2016

Login to get access

Abstract

Background

Autism spectrum disorders (ASDs) represent a heterogeneous group of disorders with a wide range of behavioral impairments including social and communication deficits. Apart from these core symptoms, a significant number of ASD individuals display higher levels of anxiety, and some studies indicate that a subset of ASD individuals have a reduced ability to be fear conditioned. Deciphering the molecular basis of ASD has been considerably challenging and it currently remains poorly understood. In this study we examined the molecular basis of autism-like impairments in an environmentally induced animal model of ASD, where pregnant rats are exposed to the known teratogen, valproic acid (VPA), on day 12.5 of gestation and the subsequent progeny exhibit ASD-like symptoms. We focused our analysis on the basal and lateral nucleus of the amygdala (BLA), a region of the brain found to be associated with ASD pathology.

Methods

We performed whole genome gene expression analysis on the BLA using DNA microarrays to examine differences in gene expression within the amygdala of VPA-exposed animals. We validated one VPA-dysregulated candidate gene (Homer1a) using both quantitative PCR (qRT-PCR) and western blot. Finally, we overexpressed Homer1a within the basal and lateral amygdala of naïve animals utilizing adeno-associated viruses (AAV) and subsequently examined these animals in a battery of behavioral tests associated with ASD, including auditory fear conditioning, social interaction and open field.

Results

Our microarray data indicated that Homer1a was one of the genes which exhibited a significant upregulation within the amygdala. We observed an increase in Homer1a messenger RNA (mRNA) and protein in multiple cohorts of VPA-exposed animals indicating that dysregulation of Homer1a levels might underlie some of the symptoms exhibited by VPA-exposed animals. To test this hypothesis, we overexpressed Homer1a within BLA neurons utilizing a viral-mediated approach and found that overexpression of Homer1a impaired auditory fear conditioning and reduced social interaction, while having no influence on open-field behavior.

Conclusions

This study indicates that dysregulation of amygdala Homer1a might contribute to some autism-like symptoms induced by VPA exposure. These findings are interesting in part because Homer1a influences the functioning of Shank3, metabotropic glutamate receptors (mGluR5), and Homer1, and these proteins have previously been associated with ASD, indicating that these differing models of ASD may have a similar molecular basis.
Appendix
Available only for authorised users
Literature
1.
go back to reference Cohen S, Conduit R, Lockley SW, Rajaratnam SM, Cornish KM. The relationship between sleep and behavior in autism spectrum disorder (ASD): a review. J Neurodev Disord. 2014;6:44.PubMedCentralPubMedCrossRef Cohen S, Conduit R, Lockley SW, Rajaratnam SM, Cornish KM. The relationship between sleep and behavior in autism spectrum disorder (ASD): a review. J Neurodev Disord. 2014;6:44.PubMedCentralPubMedCrossRef
2.
3.
go back to reference Simonoff E, Pickles A, Charman T, Chandler S, Loucas T, Baird G. Psychiatric disorders in children with autism spectrum disorders: prevalence, comorbidity, and associated factors in a population-derived sample. J Am Acad Child Adolesc Psychiatry. 2008;47:921–9.PubMedCrossRef Simonoff E, Pickles A, Charman T, Chandler S, Loucas T, Baird G. Psychiatric disorders in children with autism spectrum disorders: prevalence, comorbidity, and associated factors in a population-derived sample. J Am Acad Child Adolesc Psychiatry. 2008;47:921–9.PubMedCrossRef
4.
go back to reference Wolff JJ, Symons FJ. An evaluation of multi-component exposure treatment of needle phobia in an adult with autism and intellectual disability. J Appl Res Intellect Disabil. 2013;26:344–8.PubMedCrossRef Wolff JJ, Symons FJ. An evaluation of multi-component exposure treatment of needle phobia in an adult with autism and intellectual disability. J Appl Res Intellect Disabil. 2013;26:344–8.PubMedCrossRef
5.
go back to reference Evans DW, Canavera K, Kleinpeter FL, Maccubbin E, Taga K. The fears, phobias and anxieties of children with autism spectrum disorders and down syndrome: comparisons with developmentally and chronologically age matched children. Child Psychiatry Hum Dev. 2005;36:3–26.PubMedCrossRef Evans DW, Canavera K, Kleinpeter FL, Maccubbin E, Taga K. The fears, phobias and anxieties of children with autism spectrum disorders and down syndrome: comparisons with developmentally and chronologically age matched children. Child Psychiatry Hum Dev. 2005;36:3–26.PubMedCrossRef
6.
go back to reference Gaigg SB, Bowler DM. Differential fear conditioning in Asperger’s syndrome: implications for an amygdala theory of autism. Neuropsychologia. 2007;45:2125–34.PubMedCrossRef Gaigg SB, Bowler DM. Differential fear conditioning in Asperger’s syndrome: implications for an amygdala theory of autism. Neuropsychologia. 2007;45:2125–34.PubMedCrossRef
7.
go back to reference Amaral DG, Corbett BA. The amygdala, autism and anxiety. Novartis Found Symp. 2003;251:177–87. discussion 187–197, 281–197.PubMedCrossRef Amaral DG, Corbett BA. The amygdala, autism and anxiety. Novartis Found Symp. 2003;251:177–87. discussion 187–197, 281–197.PubMedCrossRef
8.
go back to reference Ashwin C, Baron-Cohen S, Wheelwright S, O'Riordan M, Bullmore ET. Differential activation of the amygdala and the 'social brain' during fearful face-processing in Asperger syndrome. Neuropsychologia. 2007;45:2–14.PubMedCrossRef Ashwin C, Baron-Cohen S, Wheelwright S, O'Riordan M, Bullmore ET. Differential activation of the amygdala and the 'social brain' during fearful face-processing in Asperger syndrome. Neuropsychologia. 2007;45:2–14.PubMedCrossRef
9.
go back to reference Hessl D, Rivera S, Koldewyn K, Cordeiro L, Adams J, Tassone F, et al. Amygdala dysfunction in men with the fragile X premutation. Brain. 2007;130:404–16.PubMedCrossRef Hessl D, Rivera S, Koldewyn K, Cordeiro L, Adams J, Tassone F, et al. Amygdala dysfunction in men with the fragile X premutation. Brain. 2007;130:404–16.PubMedCrossRef
10.
go back to reference Schultz RT. Developmental deficits in social perception in autism: the role of the amygdala and fusiform face area. Int J Dev Neurosci. 2005;23:125–41.PubMedCrossRef Schultz RT. Developmental deficits in social perception in autism: the role of the amygdala and fusiform face area. Int J Dev Neurosci. 2005;23:125–41.PubMedCrossRef
11.
go back to reference Schumann CM, Barnes CC, Lord C, Courchesne E. Amygdala enlargement in toddlers with autism related to severity of social and communication impairments. Biol Psychiatry. 2009;66:942–9.PubMedCentralPubMedCrossRef Schumann CM, Barnes CC, Lord C, Courchesne E. Amygdala enlargement in toddlers with autism related to severity of social and communication impairments. Biol Psychiatry. 2009;66:942–9.PubMedCentralPubMedCrossRef
12.
go back to reference Sweeten TL, Posey DJ, Shekhar A, McDougle CJ. The amygdala and related structures in the pathophysiology of autism. Pharmacol Biochem Behav. 2002;71:449–55.PubMedCrossRef Sweeten TL, Posey DJ, Shekhar A, McDougle CJ. The amygdala and related structures in the pathophysiology of autism. Pharmacol Biochem Behav. 2002;71:449–55.PubMedCrossRef
13.
go back to reference Preissler MA. Associative learning of pictures and words by low-functioning children with autism. Autism. 2008;12:231–48.PubMedCrossRef Preissler MA. Associative learning of pictures and words by low-functioning children with autism. Autism. 2008;12:231–48.PubMedCrossRef
14.
go back to reference Michaelson JJ, Shi Y, Gujral M, Zheng H, Malhotra D, Jin X, et al. Whole-genome sequencing in autism identifies hot spots for de novo germline mutation. Cell. 2012;151:1431–42.PubMedCentralPubMedCrossRef Michaelson JJ, Shi Y, Gujral M, Zheng H, Malhotra D, Jin X, et al. Whole-genome sequencing in autism identifies hot spots for de novo germline mutation. Cell. 2012;151:1431–42.PubMedCentralPubMedCrossRef
15.
go back to reference Hallmayer J, Cleveland S, Torres A, Phillips J, Cohen B, Torigoe T, et al. Genetic heritability and shared environmental factors among twin pairs with autism. Arch Gen Psychiatry. 2011;68:1095–102.PubMedCentralPubMedCrossRef Hallmayer J, Cleveland S, Torres A, Phillips J, Cohen B, Torigoe T, et al. Genetic heritability and shared environmental factors among twin pairs with autism. Arch Gen Psychiatry. 2011;68:1095–102.PubMedCentralPubMedCrossRef
16.
go back to reference Bailey A, Le Couteur A, Gottesman I, Bolton P, Simonoff E, Yuzda E, et al. Autism as a strongly genetic disorder: evidence from a British twin study. Psychol Med. 1995;25:63–77.PubMedCrossRef Bailey A, Le Couteur A, Gottesman I, Bolton P, Simonoff E, Yuzda E, et al. Autism as a strongly genetic disorder: evidence from a British twin study. Psychol Med. 1995;25:63–77.PubMedCrossRef
17.
go back to reference Toma C, Torrico B, Hervas A, Valdes-Mas R, Tristan-Noguero A, Padillo V, et al. Exome sequencing in multiplex autism families suggests a major role for heterozygous truncating mutations. Mol Psychiatry. 2014;19:784–90.PubMedCrossRef Toma C, Torrico B, Hervas A, Valdes-Mas R, Tristan-Noguero A, Padillo V, et al. Exome sequencing in multiplex autism families suggests a major role for heterozygous truncating mutations. Mol Psychiatry. 2014;19:784–90.PubMedCrossRef
18.
go back to reference Egger G, Roetzer KM, Noor A, Lionel AC, Mahmood H, Schwarzbraun T, et al. Identification of risk genes for autism spectrum disorder through copy number variation analysis in Austrian families. Neurogenetics. 2014;15:117–27.PubMedCrossRef Egger G, Roetzer KM, Noor A, Lionel AC, Mahmood H, Schwarzbraun T, et al. Identification of risk genes for autism spectrum disorder through copy number variation analysis in Austrian families. Neurogenetics. 2014;15:117–27.PubMedCrossRef
19.
go back to reference Nava C, Keren B, Mignot C, Rastetter A, Chantot-Bastaraud S, Faudet A, et al. Prospective diagnostic analysis of copy number variants using SNP microarrays in individuals with autism spectrum disorders. Eur J Hum Genet. 2014;22:71–8.PubMedCentralPubMedCrossRef Nava C, Keren B, Mignot C, Rastetter A, Chantot-Bastaraud S, Faudet A, et al. Prospective diagnostic analysis of copy number variants using SNP microarrays in individuals with autism spectrum disorders. Eur J Hum Genet. 2014;22:71–8.PubMedCentralPubMedCrossRef
20.
go back to reference Shishido E, Aleksic B, Ozaki N. Copy-number variation in the pathogenesis of autism spectrum disorder. Psychiatry Clin Neurosci. 2014;68:85–95.PubMedCrossRef Shishido E, Aleksic B, Ozaki N. Copy-number variation in the pathogenesis of autism spectrum disorder. Psychiatry Clin Neurosci. 2014;68:85–95.PubMedCrossRef
21.
go back to reference Yuen RK, Thiruvahindrapuram B, Merico D, Walker S, Tammimies K, Hoang N, et al. Whole-genome sequencing of quartet families with autism spectrum disorder. Nat Med. 2015;21:185–91.PubMedCrossRef Yuen RK, Thiruvahindrapuram B, Merico D, Walker S, Tammimies K, Hoang N, et al. Whole-genome sequencing of quartet families with autism spectrum disorder. Nat Med. 2015;21:185–91.PubMedCrossRef
22.
go back to reference Rasalam AD, Hailey H, Williams JH, Moore SJ, Turnpenny PD, Lloyd DJ, et al. Characteristics of fetal anticonvulsant syndrome associated autistic disorder. Dev Med Child Neurol. 2005;47:551–5.PubMedCrossRef Rasalam AD, Hailey H, Williams JH, Moore SJ, Turnpenny PD, Lloyd DJ, et al. Characteristics of fetal anticonvulsant syndrome associated autistic disorder. Dev Med Child Neurol. 2005;47:551–5.PubMedCrossRef
23.
go back to reference Moore SJ, Turnpenny P, Quinn A, Glover S, Lloyd DJ, Montgomery T, et al. A clinical study of 57 children with fetal anticonvulsant syndromes. J Med Genet. 2000;37:489–97.PubMedCentralPubMedCrossRef Moore SJ, Turnpenny P, Quinn A, Glover S, Lloyd DJ, Montgomery T, et al. A clinical study of 57 children with fetal anticonvulsant syndromes. J Med Genet. 2000;37:489–97.PubMedCentralPubMedCrossRef
24.
go back to reference Christianson AL, Chesler N, Kromberg JG. Fetal valproate syndrome: clinical and neuro-developmental features in two sibling pairs. Dev Med Child Neurol. 1994;36:361–9.PubMedCrossRef Christianson AL, Chesler N, Kromberg JG. Fetal valproate syndrome: clinical and neuro-developmental features in two sibling pairs. Dev Med Child Neurol. 1994;36:361–9.PubMedCrossRef
25.
go back to reference Kim KC, Kim P, Go HS, Choi CS, Yang SI, Cheong JH, et al. The critical period of valproate exposure to induce autistic symptoms in Sprague–Dawley rats. Toxicol Lett. 2011;201:137–42.PubMedCrossRef Kim KC, Kim P, Go HS, Choi CS, Yang SI, Cheong JH, et al. The critical period of valproate exposure to induce autistic symptoms in Sprague–Dawley rats. Toxicol Lett. 2011;201:137–42.PubMedCrossRef
26.
go back to reference Rodier PM, Ingram JL, Tisdale B, Nelson S, Romano J. Embryological origin for autism: developmental anomalies of the cranial nerve motor nuclei. J Comp Neurol. 1996;370:247–61.PubMedCrossRef Rodier PM, Ingram JL, Tisdale B, Nelson S, Romano J. Embryological origin for autism: developmental anomalies of the cranial nerve motor nuclei. J Comp Neurol. 1996;370:247–61.PubMedCrossRef
27.
28.
go back to reference Vorhees CV. Behavioral teratogenicity of valproic acid: selective effects on behavior after prenatal exposure to rats. Psychopharmacology (Berl). 1987;92:173–9.CrossRef Vorhees CV. Behavioral teratogenicity of valproic acid: selective effects on behavior after prenatal exposure to rats. Psychopharmacology (Berl). 1987;92:173–9.CrossRef
29.
go back to reference Kemper TL, Bauman M. Neuropathology of infantile autism. J Neuropathol Exp Neurol. 1998;57:645–52.PubMedCrossRef Kemper TL, Bauman M. Neuropathology of infantile autism. J Neuropathol Exp Neurol. 1998;57:645–52.PubMedCrossRef
30.
go back to reference Schneider T, Przewlocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology. 2005;30:80–9.PubMedCrossRef Schneider T, Przewlocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology. 2005;30:80–9.PubMedCrossRef
31.
go back to reference Schneider T, Ziolkowska B, Gieryk A, Tyminska A, Przewlocki R. Prenatal exposure to valproic acid disturbs the enkephalinergic system functioning, basal hedonic tone, and emotional responses in an animal model of autism. Psychopharmacology (Berl). 2007;193:547–55.CrossRef Schneider T, Ziolkowska B, Gieryk A, Tyminska A, Przewlocki R. Prenatal exposure to valproic acid disturbs the enkephalinergic system functioning, basal hedonic tone, and emotional responses in an animal model of autism. Psychopharmacology (Berl). 2007;193:547–55.CrossRef
32.
go back to reference Banerjee A, Garcia-Oscos F, Roychowdhury S, Galindo LC, Hall S, Kilgard MP, et al. Impairment of cortical GABAergic synaptic transmission in an environmental rat model of autism. Int J Neuropsychopharmacol. 2013;16:1309–18.PubMedCentralPubMedCrossRef Banerjee A, Garcia-Oscos F, Roychowdhury S, Galindo LC, Hall S, Kilgard MP, et al. Impairment of cortical GABAergic synaptic transmission in an environmental rat model of autism. Int J Neuropsychopharmacol. 2013;16:1309–18.PubMedCentralPubMedCrossRef
33.
go back to reference Markram K, Rinaldi T, La Mendola D, Sandi C, Markram H. Abnormal fear conditioning and amygdala processing in an animal model of autism. Neuropsychopharmacology. 2008;33:901–12.PubMedCrossRef Markram K, Rinaldi T, La Mendola D, Sandi C, Markram H. Abnormal fear conditioning and amygdala processing in an animal model of autism. Neuropsychopharmacology. 2008;33:901–12.PubMedCrossRef
34.
go back to reference Lin HC, Gean PW, Wang CC, Chan YH, Chen PS. The amygdala excitatory/inhibitory balance in a valproate-induced rat autism model. PLoS One. 2013;8:e55248.PubMedCentralPubMedCrossRef Lin HC, Gean PW, Wang CC, Chan YH, Chen PS. The amygdala excitatory/inhibitory balance in a valproate-induced rat autism model. PLoS One. 2013;8:e55248.PubMedCentralPubMedCrossRef
35.
go back to reference Wang CC, Lin HC, Chan YH, Gean PW, Yang YK, Chen PS. 5-HT1A-receptor agonist modified amygdala activity and amygdala-associated social behavior in a valproate-induced rat autism model. Int J Neuropsychopharmacol. 2013;16:2027–39.PubMedCrossRef Wang CC, Lin HC, Chan YH, Gean PW, Yang YK, Chen PS. 5-HT1A-receptor agonist modified amygdala activity and amygdala-associated social behavior in a valproate-induced rat autism model. Int J Neuropsychopharmacol. 2013;16:2027–39.PubMedCrossRef
36.
go back to reference Banerjee A, Engineer CT, Sauls BL, Morales AA, Kilgard MP, Ploski JE. Abnormal emotional learning in a rat model of autism exposed to valproic acid in utero. Front Behav Neurosci. 2014;8:387.PubMedCentralPubMed Banerjee A, Engineer CT, Sauls BL, Morales AA, Kilgard MP, Ploski JE. Abnormal emotional learning in a rat model of autism exposed to valproic acid in utero. Front Behav Neurosci. 2014;8:387.PubMedCentralPubMed
37.
38.
go back to reference Ronesi JA, Collins KA, Hays SA, Tsai NP, Guo W, Birnbaum SG, et al. Disrupted Homer scaffolds mediate abnormal mGluR5 function in a mouse model of fragile X syndrome. Nat Neurosci. 2012;15:431–40. S431.PubMedCentralPubMedCrossRef Ronesi JA, Collins KA, Hays SA, Tsai NP, Guo W, Birnbaum SG, et al. Disrupted Homer scaffolds mediate abnormal mGluR5 function in a mouse model of fragile X syndrome. Nat Neurosci. 2012;15:431–40. S431.PubMedCentralPubMedCrossRef
39.
go back to reference Calfa G, Percy AK, Pozzo-Miller L. Experimental models of Rett syndrome based on Mecp2 dysfunction. Exp Biol Med (Maywood). 2011;236:3–19.CrossRef Calfa G, Percy AK, Pozzo-Miller L. Experimental models of Rett syndrome based on Mecp2 dysfunction. Exp Biol Med (Maywood). 2011;236:3–19.CrossRef
40.
go back to reference Greer PL, Hanayama R, Bloodgood BL, Mardinly AR, Lipton DM, Flavell SW, et al. The Angelman syndrome protein Ube3A regulates synapse development by ubiquitinating arc. Cell. 2010;140:704–16.PubMedCentralPubMedCrossRef Greer PL, Hanayama R, Bloodgood BL, Mardinly AR, Lipton DM, Flavell SW, et al. The Angelman syndrome protein Ube3A regulates synapse development by ubiquitinating arc. Cell. 2010;140:704–16.PubMedCentralPubMedCrossRef
41.
go back to reference Kirschstein T. Synaptic plasticity and learning in animal models of tuberous sclerosis complex. Neural Plast. 2012;2012:279834.PubMedCentralPubMed Kirschstein T. Synaptic plasticity and learning in animal models of tuberous sclerosis complex. Neural Plast. 2012;2012:279834.PubMedCentralPubMed
42.
go back to reference Durand CM, Betancur C, Boeckers TM, Bockmann J, Chaste P, Fauchereau F, et al. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat Genet. 2007;39:25–7.PubMedCentralPubMedCrossRef Durand CM, Betancur C, Boeckers TM, Bockmann J, Chaste P, Fauchereau F, et al. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat Genet. 2007;39:25–7.PubMedCentralPubMedCrossRef
43.
go back to reference Blundell J, Blaiss CA, Etherton MR, Espinosa F, Tabuchi K, Walz C, et al. Neuroligin-1 deletion results in impaired spatial memory and increased repetitive behavior. J Neurosci. 2010;30:2115–2129.PubMedCentralPubMedCrossRef Blundell J, Blaiss CA, Etherton MR, Espinosa F, Tabuchi K, Walz C, et al. Neuroligin-1 deletion results in impaired spatial memory and increased repetitive behavior. J Neurosci. 2010;30:2115–2129.PubMedCentralPubMedCrossRef
44.
go back to reference Hung AY, Futai K, Sala C, Valtschanoff JG, Ryu J, Woodworth MA, et al. Smaller dendritic spines, weaker synaptic transmission, but enhanced spatial learning in mice lacking Shank1. J Neurosci. 2008;28:1697–708.PubMedCentralPubMedCrossRef Hung AY, Futai K, Sala C, Valtschanoff JG, Ryu J, Woodworth MA, et al. Smaller dendritic spines, weaker synaptic transmission, but enhanced spatial learning in mice lacking Shank1. J Neurosci. 2008;28:1697–708.PubMedCentralPubMedCrossRef
45.
go back to reference Adachi M, Autry AE, Covington 3rd HE, Monteggia LM. MeCP2-mediated transcription repression in the basolateral amygdala may underlie heightened anxiety in a mouse model of Rett syndrome. J Neurosci. 2009;29:4218–27.PubMedCentralPubMedCrossRef Adachi M, Autry AE, Covington 3rd HE, Monteggia LM. MeCP2-mediated transcription repression in the basolateral amygdala may underlie heightened anxiety in a mouse model of Rett syndrome. J Neurosci. 2009;29:4218–27.PubMedCentralPubMedCrossRef
46.
go back to reference Kim J, Jung SY, Lee YK, Park S, Choi JS, Lee CJ, et al. Neuroligin-1 is required for normal expression of LTP and associative fear memory in the amygdala of adult animals. Proc Natl Acad Sci U S A. 2008;105:9087–92.PubMedCentralPubMedCrossRef Kim J, Jung SY, Lee YK, Park S, Choi JS, Lee CJ, et al. Neuroligin-1 is required for normal expression of LTP and associative fear memory in the amygdala of adult animals. Proc Natl Acad Sci U S A. 2008;105:9087–92.PubMedCentralPubMedCrossRef
47.
go back to reference Choi YB, Li HL, Kassabov SR, Jin I, Puthanveettil SV, Karl KA, et al. Neurexin-neuroligin transsynaptic interaction mediates learning-related synaptic remodeling and long-term facilitation in aplysia. Neuron. 2011;70:468–81.PubMedCentralPubMedCrossRef Choi YB, Li HL, Kassabov SR, Jin I, Puthanveettil SV, Karl KA, et al. Neurexin-neuroligin transsynaptic interaction mediates learning-related synaptic remodeling and long-term facilitation in aplysia. Neuron. 2011;70:468–81.PubMedCentralPubMedCrossRef
48.
go back to reference Ango F, Pin JP, Tu JC, Xiao B, Worley PF, Bockaert J, et al. Dendritic and axonal targeting of type 5 metabotropic glutamate receptor is regulated by homer1 proteins and neuronal excitation. J Neurosci. 2000;20:8710–6.PubMed Ango F, Pin JP, Tu JC, Xiao B, Worley PF, Bockaert J, et al. Dendritic and axonal targeting of type 5 metabotropic glutamate receptor is regulated by homer1 proteins and neuronal excitation. J Neurosci. 2000;20:8710–6.PubMed
49.
go back to reference Tu JC, Xiao B, Naisbitt S, Yuan JP, Petralia RS, Brakeman P, et al. Coupling of mGluR/Homer and PSD-95 complexes by the Shank family of postsynaptic density proteins. Neuron. 1999;23:583–92.PubMedCrossRef Tu JC, Xiao B, Naisbitt S, Yuan JP, Petralia RS, Brakeman P, et al. Coupling of mGluR/Homer and PSD-95 complexes by the Shank family of postsynaptic density proteins. Neuron. 1999;23:583–92.PubMedCrossRef
50.
go back to reference Xiao B, Tu JC, Petralia RS, Yuan JP, Doan A, Breder CD, et al. Homer regulates the association of group 1 metabotropic glutamate receptors with multivalent complexes of homer-related, synaptic proteins. Neuron. 1998;21:707–16.PubMedCrossRef Xiao B, Tu JC, Petralia RS, Yuan JP, Doan A, Breder CD, et al. Homer regulates the association of group 1 metabotropic glutamate receptors with multivalent complexes of homer-related, synaptic proteins. Neuron. 1998;21:707–16.PubMedCrossRef
51.
go back to reference Cavarsan CF, Tescarollo F, Tesone-Coelho C, Morais RL, Motta FL, Blanco MM, et al. Pilocarpine-induced status epilepticus increases Homer1a and changes mGluR5 expression. Epilepsy Res. 2012;101:253–60.PubMedCrossRef Cavarsan CF, Tescarollo F, Tesone-Coelho C, Morais RL, Motta FL, Blanco MM, et al. Pilocarpine-induced status epilepticus increases Homer1a and changes mGluR5 expression. Epilepsy Res. 2012;101:253–60.PubMedCrossRef
52.
go back to reference Obara I, Goulding SP, Hu JH, Klugmann M, Worley PF, Szumlinski KK. Nerve injury-induced changes in Homer/glutamate receptor signaling contribute to the development and maintenance of neuropathic pain. Pain. 2013;154:1932–45.PubMedCrossRef Obara I, Goulding SP, Hu JH, Klugmann M, Worley PF, Szumlinski KK. Nerve injury-induced changes in Homer/glutamate receptor signaling contribute to the development and maintenance of neuropathic pain. Pain. 2013;154:1932–45.PubMedCrossRef
53.
go back to reference O'Riordan K, Gerstein H, Hullinger R, Burger C. The role of Homer1c in metabotropic glutamate receptor-dependent long-term potentiation. Hippocampus. 2014;24:1–6.PubMedCrossRef O'Riordan K, Gerstein H, Hullinger R, Burger C. The role of Homer1c in metabotropic glutamate receptor-dependent long-term potentiation. Hippocampus. 2014;24:1–6.PubMedCrossRef
54.
go back to reference Saito H, Kimura M, Inanobe A, Ohe T, Kurachi Y. An N-terminal sequence specific for a novel Homer1 isoform controls trafficking of group I metabotropic glutamate receptor in mammalian cells. Biochem Biophys Res Commun. 2002;296:523–9.PubMedCrossRef Saito H, Kimura M, Inanobe A, Ohe T, Kurachi Y. An N-terminal sequence specific for a novel Homer1 isoform controls trafficking of group I metabotropic glutamate receptor in mammalian cells. Biochem Biophys Res Commun. 2002;296:523–9.PubMedCrossRef
55.
go back to reference Mahan AL, Mou L, Shah N, Hu JH, Worley PF, Ressler KJ. Epigenetic modulation of Homer1a transcription regulation in amygdala and hippocampus with pavlovian fear conditioning. J Neurosci. 2012;32:4651–9.PubMedCentralPubMedCrossRef Mahan AL, Mou L, Shah N, Hu JH, Worley PF, Ressler KJ. Epigenetic modulation of Homer1a transcription regulation in amygdala and hippocampus with pavlovian fear conditioning. J Neurosci. 2012;32:4651–9.PubMedCentralPubMedCrossRef
56.
go back to reference Inoue N, Nakao H, Migishima R, Hino T, Matsui M, Hayashi F, et al. Requirement of the immediate early gene vesl-1S/homer-1a for fear memory formation. Mol Brain. 2009;2:7.PubMedCentralPubMedCrossRef Inoue N, Nakao H, Migishima R, Hino T, Matsui M, Hayashi F, et al. Requirement of the immediate early gene vesl-1S/homer-1a for fear memory formation. Mol Brain. 2009;2:7.PubMedCentralPubMedCrossRef
57.
go back to reference Rozov A, Zivkovic AR, Schwarz MK. Homer1 gene products orchestrate Ca(2+)-permeable AMPA receptor distribution and LTP expression. Front Synaptic Neurosci. 2012;4:4.PubMedCentralPubMedCrossRef Rozov A, Zivkovic AR, Schwarz MK. Homer1 gene products orchestrate Ca(2+)-permeable AMPA receptor distribution and LTP expression. Front Synaptic Neurosci. 2012;4:4.PubMedCentralPubMedCrossRef
58.
go back to reference Celikel T, Marx V, Freudenberg F, Zivkovic A, Resnik E, Hasan MT, et al. Select overexpression of homer1a in dorsal hippocampus impairs spatial working memory. Front Neurosci. 2007;1:97–110.PubMedCentralPubMedCrossRef Celikel T, Marx V, Freudenberg F, Zivkovic A, Resnik E, Hasan MT, et al. Select overexpression of homer1a in dorsal hippocampus impairs spatial working memory. Front Neurosci. 2007;1:97–110.PubMedCentralPubMedCrossRef
60.
go back to reference Adolphs R, Tranel D, Damasio H, Damasio A. Impaired recognition of emotion in facial expressions following bilateral damage to the human amygdala. Nature. 1994;372:669–72.PubMedCrossRef Adolphs R, Tranel D, Damasio H, Damasio A. Impaired recognition of emotion in facial expressions following bilateral damage to the human amygdala. Nature. 1994;372:669–72.PubMedCrossRef
62.
go back to reference Davis M. Neurobiology of fear responses: the role of the amygdala. J Neuropsychiatry Clin Neurosci. 1997;9:382–402.PubMedCrossRef Davis M. Neurobiology of fear responses: the role of the amygdala. J Neuropsychiatry Clin Neurosci. 1997;9:382–402.PubMedCrossRef
63.
go back to reference Hamm AO, Weike AI. The neuropsychology of fear learning and fear regulation. Int J Psychophysiol. 2005;57:5–14.PubMedCrossRef Hamm AO, Weike AI. The neuropsychology of fear learning and fear regulation. Int J Psychophysiol. 2005;57:5–14.PubMedCrossRef
64.
go back to reference Kelleher 3rd RJ, Geigenmuller U, Hovhannisyan H, Trautman E, Pinard R, Rathmell B, et al. High-throughput sequencing of mGluR signaling pathway genes reveals enrichment of rare variants in autism. PLoS One. 2012;7:e35003.PubMedCrossRef Kelleher 3rd RJ, Geigenmuller U, Hovhannisyan H, Trautman E, Pinard R, Rathmell B, et al. High-throughput sequencing of mGluR signaling pathway genes reveals enrichment of rare variants in autism. PLoS One. 2012;7:e35003.PubMedCrossRef
65.
go back to reference Banerjee A, Engineer CT, Sauls BL, Morales AA, Kilgard MP, Ploski JE. Abnormal emotional learning in a rat model of autism exposed to valproic acid in utero. Frontiers in Behavioral Neuroscience. 2014;8:387.PubMedCentralPubMedCrossRef Banerjee A, Engineer CT, Sauls BL, Morales AA, Kilgard MP, Ploski JE. Abnormal emotional learning in a rat model of autism exposed to valproic acid in utero. Frontiers in Behavioral Neuroscience. 2014;8:387.PubMedCentralPubMedCrossRef
66.
go back to reference Shi L, Reid LH, Jones WD, Shippy R, Warrington JA, Baker SC, et al. The MicroArray Quality Control (MAQC) project shows inter- and intraplatform reproducibility of gene expression measurements. Nat Biotechnol. 2006;24:1151–61.PubMedCrossRef Shi L, Reid LH, Jones WD, Shippy R, Warrington JA, Baker SC, et al. The MicroArray Quality Control (MAQC) project shows inter- and intraplatform reproducibility of gene expression measurements. Nat Biotechnol. 2006;24:1151–61.PubMedCrossRef
67.
go back to reference Partin AC, Hosek MP, Luong JA, Lella SK, Sharma SA, Ploski JE. Amygdala nuclei critical for emotional learning exhibit unique gene expression patterns. Neurobiol Learn Mem. 2013;104:110–21.PubMedCentralPubMedCrossRef Partin AC, Hosek MP, Luong JA, Lella SK, Sharma SA, Ploski JE. Amygdala nuclei critical for emotional learning exhibit unique gene expression patterns. Neurobiol Learn Mem. 2013;104:110–21.PubMedCentralPubMedCrossRef
68.
go back to reference Garcia EP, Dowding LA, Stanton LW, Slepnev VI. Scalable transcriptional analysis routine—multiplexed quantitative real-time polymerase chain reaction platform for gene expression analysis and molecular diagnostics. J Mol Diagn. 2005;7:444–54.PubMedCentralPubMedCrossRef Garcia EP, Dowding LA, Stanton LW, Slepnev VI. Scalable transcriptional analysis routine—multiplexed quantitative real-time polymerase chain reaction platform for gene expression analysis and molecular diagnostics. J Mol Diagn. 2005;7:444–54.PubMedCentralPubMedCrossRef
69.
go back to reference Lominac KD, Oleson EB, Pava M, Klugmann M, Schwarz MK, Seeburg PH, et al. Distinct roles for different Homer1 isoforms in behaviors and associated prefrontal cortex function. J Neurosci. 2005;25:11586–94.PubMedCrossRef Lominac KD, Oleson EB, Pava M, Klugmann M, Schwarz MK, Seeburg PH, et al. Distinct roles for different Homer1 isoforms in behaviors and associated prefrontal cortex function. J Neurosci. 2005;25:11586–94.PubMedCrossRef
70.
go back to reference Klugmann M, Szumlinski KK. Targeting Homer genes using adeno-associated viral vector: lessons learned from behavioural and neurochemical studies. Behav Pharmacol. 2008;19:485–500.PubMedCrossRef Klugmann M, Szumlinski KK. Targeting Homer genes using adeno-associated viral vector: lessons learned from behavioural and neurochemical studies. Behav Pharmacol. 2008;19:485–500.PubMedCrossRef
71.
go back to reference Holehonnur R, Luong JA, Chaturvedi D, Ho A, Lella SK, Hosek MP, et al. Adeno-associated viral serotypes produce differing titers and differentially transduce neurons within the rat basal and lateral amygdala. BMC Neurosci. 2014;15:28.PubMedCentralPubMedCrossRef Holehonnur R, Luong JA, Chaturvedi D, Ho A, Lella SK, Hosek MP, et al. Adeno-associated viral serotypes produce differing titers and differentially transduce neurons within the rat basal and lateral amygdala. BMC Neurosci. 2014;15:28.PubMedCentralPubMedCrossRef
74.
go back to reference Jaholkowski P, Kiryk A, Jedynak P, Ben Abdallah NM, Knapska E, Kowalczyk A, et al. New hippocampal neurons are not obligatory for memory formation; cyclin D2 knockout mice with no adult brain neurogenesis show learning. Learn Mem. 2009;16:439–51.PubMedCrossRef Jaholkowski P, Kiryk A, Jedynak P, Ben Abdallah NM, Knapska E, Kowalczyk A, et al. New hippocampal neurons are not obligatory for memory formation; cyclin D2 knockout mice with no adult brain neurogenesis show learning. Learn Mem. 2009;16:439–51.PubMedCrossRef
75.
go back to reference Bottai D, Guzowski JF, Schwarz MK, Kang SH, Xiao B, Lanahan A, et al. Synaptic activity-induced conversion of intronic to exonic sequence in Homer 1 immediate early gene expression. J Neurosci. 2002;22:167–75.PubMed Bottai D, Guzowski JF, Schwarz MK, Kang SH, Xiao B, Lanahan A, et al. Synaptic activity-induced conversion of intronic to exonic sequence in Homer 1 immediate early gene expression. J Neurosci. 2002;22:167–75.PubMed
76.
go back to reference Tappe A, Klugmann M, Luo C, Hirlinger D, Agarwal N, Benrath J, et al. Synaptic scaffolding protein Homer1a protects against chronic inflammatory pain. Nat Med. 2006;12:677–81.PubMedCrossRef Tappe A, Klugmann M, Luo C, Hirlinger D, Agarwal N, Benrath J, et al. Synaptic scaffolding protein Homer1a protects against chronic inflammatory pain. Nat Med. 2006;12:677–81.PubMedCrossRef
77.
go back to reference Bliss-Moreau E, Moadab G, Bauman MD, Amaral DG. The impact of early amygdala damage on juvenile rhesus macaque social behavior. J Cogn Neurosci. 2013;25:2124–2140.PubMedCentralPubMedCrossRef Bliss-Moreau E, Moadab G, Bauman MD, Amaral DG. The impact of early amygdala damage on juvenile rhesus macaque social behavior. J Cogn Neurosci. 2013;25:2124–2140.PubMedCentralPubMedCrossRef
78.
go back to reference Brakeman PR, Lanahan AA, O'Brien R, Roche K, Barnes CA, Huganir RL, et al. Homer: a protein that selectively binds metabotropic glutamate receptors. Nature. 1997;386:284–8.PubMedCrossRef Brakeman PR, Lanahan AA, O'Brien R, Roche K, Barnes CA, Huganir RL, et al. Homer: a protein that selectively binds metabotropic glutamate receptors. Nature. 1997;386:284–8.PubMedCrossRef
79.
go back to reference Tu JC, Xiao B, Yuan JP, Lanahan AA, Leoffert K, Li M, et al. Homer binds a novel proline-rich motif and links group 1 metabotropic glutamate receptors with IP3 receptors. Neuron. 1998;21:717–26.PubMedCrossRef Tu JC, Xiao B, Yuan JP, Lanahan AA, Leoffert K, Li M, et al. Homer binds a novel proline-rich motif and links group 1 metabotropic glutamate receptors with IP3 receptors. Neuron. 1998;21:717–26.PubMedCrossRef
80.
go back to reference Gross C, Raj N, Molinaro G, Allen AG, Whyte AJ, Gibson JR, et al. Selective role of the catalytic PI3K subunit p110beta in impaired higher order cognition in fragile X syndrome. Cell Rep. 2015;11:681–8.PubMedCrossRef Gross C, Raj N, Molinaro G, Allen AG, Whyte AJ, Gibson JR, et al. Selective role of the catalytic PI3K subunit p110beta in impaired higher order cognition in fragile X syndrome. Cell Rep. 2015;11:681–8.PubMedCrossRef
81.
go back to reference Anagnostaras SG, Josselyn SA, Frankland PW, Silva AJ. Computer-assisted behavioral assessment of Pavlovian fear conditioning in mice. Learn Mem. 2000;7:58–72.PubMedCentralPubMedCrossRef Anagnostaras SG, Josselyn SA, Frankland PW, Silva AJ. Computer-assisted behavioral assessment of Pavlovian fear conditioning in mice. Learn Mem. 2000;7:58–72.PubMedCentralPubMedCrossRef
82.
go back to reference Ghasemzadeh MB, Permenter LK, Lake R, Worley PF, Kalivas PW. Homer1 proteins and AMPA receptors modulate cocaine-induced behavioural plasticity. Eur J Neurosci. 2003;18:1645–51.PubMedCrossRef Ghasemzadeh MB, Permenter LK, Lake R, Worley PF, Kalivas PW. Homer1 proteins and AMPA receptors modulate cocaine-induced behavioural plasticity. Eur J Neurosci. 2003;18:1645–51.PubMedCrossRef
83.
go back to reference Siddoway B, Hou H, Xia H. Molecular mechanisms of homeostatic synaptic downscaling. Neuropharmacology. 2014;78:38–44.PubMedCrossRef Siddoway B, Hou H, Xia H. Molecular mechanisms of homeostatic synaptic downscaling. Neuropharmacology. 2014;78:38–44.PubMedCrossRef
84.
go back to reference Sala C, Futai K, Yamamoto K, Worley PF, Hayashi Y, Sheng M. Inhibition of dendritic spine morphogenesis and synaptic transmission by activity-inducible protein Homer1a. J Neurosci. 2003;23:6327–37.PubMed Sala C, Futai K, Yamamoto K, Worley PF, Hayashi Y, Sheng M. Inhibition of dendritic spine morphogenesis and synaptic transmission by activity-inducible protein Homer1a. J Neurosci. 2003;23:6327–37.PubMed
85.
go back to reference Fatemi SH, Folsom TD, Kneeland RE, Yousefi MK, Liesch SB, Thuras PD. Impairment of fragile X mental retardation protein-metabotropic glutamate receptor 5 signaling and its downstream cognates ras-related C3 botulinum toxin substrate 1, amyloid beta A4 precursor protein, striatal-enriched protein tyrosine phosphatase, and homer 1, in autism: a postmortem study in cerebellar vermis and superior frontal cortex. Mol Autism. 2013;4:21.PubMedCentralPubMedCrossRef Fatemi SH, Folsom TD, Kneeland RE, Yousefi MK, Liesch SB, Thuras PD. Impairment of fragile X mental retardation protein-metabotropic glutamate receptor 5 signaling and its downstream cognates ras-related C3 botulinum toxin substrate 1, amyloid beta A4 precursor protein, striatal-enriched protein tyrosine phosphatase, and homer 1, in autism: a postmortem study in cerebellar vermis and superior frontal cortex. Mol Autism. 2013;4:21.PubMedCentralPubMedCrossRef
86.
go back to reference Pignatelli M, Piccinin S, Molinaro G, Di Menna L, Riozzi B, Cannella M, et al. Changes in mGlu5 receptor-dependent synaptic plasticity and coupling to homer proteins in the hippocampus of Ube3A hemizygous mice modeling angelman syndrome. J Neurosci. 2014;34:4558–66.PubMedCrossRef Pignatelli M, Piccinin S, Molinaro G, Di Menna L, Riozzi B, Cannella M, et al. Changes in mGlu5 receptor-dependent synaptic plasticity and coupling to homer proteins in the hippocampus of Ube3A hemizygous mice modeling angelman syndrome. J Neurosci. 2014;34:4558–66.PubMedCrossRef
87.
go back to reference Prather MD, Lavenex P, Mauldin-Jourdain ML, Mason WA, Capitanio JP, Mendoza SP, et al. Increased social fear and decreased fear of objects in monkeys with neonatal amygdala lesions. Neuroscience. 2001;106:653–8.PubMedCrossRef Prather MD, Lavenex P, Mauldin-Jourdain ML, Mason WA, Capitanio JP, Mendoza SP, et al. Increased social fear and decreased fear of objects in monkeys with neonatal amygdala lesions. Neuroscience. 2001;106:653–8.PubMedCrossRef
89.
go back to reference Zeng QY, Fan TT, Zhu P, He RQ, Bao YX, Zheng RY, et al. Comparative long-term effectiveness of a monotherapy with five antiepileptic drugs for focal epilepsy in adult patients: a prospective cohort study. PLoS One. 2015;10:e0131566.PubMedCentralPubMedCrossRef Zeng QY, Fan TT, Zhu P, He RQ, Bao YX, Zheng RY, et al. Comparative long-term effectiveness of a monotherapy with five antiepileptic drugs for focal epilepsy in adult patients: a prospective cohort study. PLoS One. 2015;10:e0131566.PubMedCentralPubMedCrossRef
90.
go back to reference Clemens B, Puskas S, Besenyei M, Kovacs NZ, Spisak T, Kis SA, et al. Valproate treatment normalizes EEG functional connectivity in successfully treated idiopathic generalized epilepsy patients. Epilepsy Res. 2014;108:1896–903.PubMedCrossRef Clemens B, Puskas S, Besenyei M, Kovacs NZ, Spisak T, Kis SA, et al. Valproate treatment normalizes EEG functional connectivity in successfully treated idiopathic generalized epilepsy patients. Epilepsy Res. 2014;108:1896–903.PubMedCrossRef
91.
go back to reference Redecker J, Wittstock M, Benecke R, Rosche J. Comparison of the effectiveness of four antiepileptic drugs in the treatment of status epilepticus according to four different efficacy criteria. Epilepsy Behav. 2015;49:341–354.CrossRef Redecker J, Wittstock M, Benecke R, Rosche J. Comparison of the effectiveness of four antiepileptic drugs in the treatment of status epilepticus according to four different efficacy criteria. Epilepsy Behav. 2015;49:341–354.CrossRef
92.
93.
go back to reference Miyazaki K, Narita N, Narita M. Maternal administration of thalidomide or valproic acid causes abnormal serotonergic neurons in the offspring: implication for pathogenesis of autism. Int J Dev Neurosci. 2005;23:287–97.PubMedCrossRef Miyazaki K, Narita N, Narita M. Maternal administration of thalidomide or valproic acid causes abnormal serotonergic neurons in the offspring: implication for pathogenesis of autism. Int J Dev Neurosci. 2005;23:287–97.PubMedCrossRef
94.
go back to reference Kwan P, Sills GJ, Brodie MJ. The mechanisms of action of commonly used antiepileptic drugs. Pharmacol Ther. 2001;90:21–34.PubMedCrossRef Kwan P, Sills GJ, Brodie MJ. The mechanisms of action of commonly used antiepileptic drugs. Pharmacol Ther. 2001;90:21–34.PubMedCrossRef
95.
go back to reference Gould TD, Quiroz JA, Singh J, Zarate CA, Manji HK. Emerging experimental therapeutics for bipolar disorder: insights from the molecular and cellular actions of current mood stabilizers. Mol Psychiatry. 2004;9:734–55.PubMedCrossRef Gould TD, Quiroz JA, Singh J, Zarate CA, Manji HK. Emerging experimental therapeutics for bipolar disorder: insights from the molecular and cellular actions of current mood stabilizers. Mol Psychiatry. 2004;9:734–55.PubMedCrossRef
96.
go back to reference Jergil M, Kultima K, Gustafson AL, Dencker L, Stigson M. Valproic acid-induced deregulation in vitro of genes associated in vivo with neural tube defects. Toxicol Sci. 2009;108:132–48.PubMedCrossRef Jergil M, Kultima K, Gustafson AL, Dencker L, Stigson M. Valproic acid-induced deregulation in vitro of genes associated in vivo with neural tube defects. Toxicol Sci. 2009;108:132–48.PubMedCrossRef
97.
go back to reference Kultima K, Jergil M, Salter H, Gustafson AL, Dencker L, Stigson M. Early transcriptional responses in mouse embryos as a basis for selection of molecular markers predictive of valproic acid teratogenicity. Reprod Toxicol. 2010;30:457–68.PubMedCrossRef Kultima K, Jergil M, Salter H, Gustafson AL, Dencker L, Stigson M. Early transcriptional responses in mouse embryos as a basis for selection of molecular markers predictive of valproic acid teratogenicity. Reprod Toxicol. 2010;30:457–68.PubMedCrossRef
98.
go back to reference Cohen OS, Varlinskaya EI, Wilson CA, Glatt SJ, Mooney SM. Acute prenatal exposure to a moderate dose of valproic acid increases social behavior and alters gene expression in rats. Int J Dev Neurosci. 2013;31:740–50.PubMedCrossRef Cohen OS, Varlinskaya EI, Wilson CA, Glatt SJ, Mooney SM. Acute prenatal exposure to a moderate dose of valproic acid increases social behavior and alters gene expression in rats. Int J Dev Neurosci. 2013;31:740–50.PubMedCrossRef
99.
go back to reference Oguchi-Katayama A, Monma A, Sekino Y, Moriguchi T, Sato K. Comparative gene expression analysis of the amygdala in autistic rat models produced by pre- and post-natal exposures to valproic acid. J Toxicol Sci. 2013;38:391–402.PubMedCrossRef Oguchi-Katayama A, Monma A, Sekino Y, Moriguchi T, Sato K. Comparative gene expression analysis of the amygdala in autistic rat models produced by pre- and post-natal exposures to valproic acid. J Toxicol Sci. 2013;38:391–402.PubMedCrossRef
100.
go back to reference Roullet FI, Wollaston L, Decatanzaro D, Foster JA. Behavioral and molecular changes in the mouse in response to prenatal exposure to the anti-epileptic drug valproic acid. Neuroscience. 2010;170:514–22.PubMedCrossRef Roullet FI, Wollaston L, Decatanzaro D, Foster JA. Behavioral and molecular changes in the mouse in response to prenatal exposure to the anti-epileptic drug valproic acid. Neuroscience. 2010;170:514–22.PubMedCrossRef
101.
go back to reference Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem. 2001;276:36734–41.PubMedCrossRef Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem. 2001;276:36734–41.PubMedCrossRef
102.
go back to reference Eyal S, Yagen B, Sobol E, Altschuler Y, Shmuel M, Bialer M. The activity of antiepileptic drugs as histone deacetylase inhibitors. Epilepsia. 2004;45:737–44.PubMedCrossRef Eyal S, Yagen B, Sobol E, Altschuler Y, Shmuel M, Bialer M. The activity of antiepileptic drugs as histone deacetylase inhibitors. Epilepsia. 2004;45:737–44.PubMedCrossRef
103.
go back to reference Haberland M, Montgomery RL, Olson EN. The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet. 2009;10:32–42.PubMedCentralPubMedCrossRef Haberland M, Montgomery RL, Olson EN. The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet. 2009;10:32–42.PubMedCentralPubMedCrossRef
104.
go back to reference Montgomery RL, Hsieh J, Barbosa AC, Richardson JA, Olson EN. Histone deacetylases 1 and 2 control the progression of neural precursors to neurons during brain development. Proc Natl Acad Sci U S A. 2009;106:7876–81.PubMedCentralPubMedCrossRef Montgomery RL, Hsieh J, Barbosa AC, Richardson JA, Olson EN. Histone deacetylases 1 and 2 control the progression of neural precursors to neurons during brain development. Proc Natl Acad Sci U S A. 2009;106:7876–81.PubMedCentralPubMedCrossRef
105.
go back to reference Shaked M, Weissmuller K, Svoboda H, Hortschansky P, Nishino N, Wolfl S, et al. Histone deacetylases control neurogenesis in embryonic brain by inhibition of BMP2/4 signaling. PLoS One. 2008;3:e2668.PubMedCentralPubMedCrossRef Shaked M, Weissmuller K, Svoboda H, Hortschansky P, Nishino N, Wolfl S, et al. Histone deacetylases control neurogenesis in embryonic brain by inhibition of BMP2/4 signaling. PLoS One. 2008;3:e2668.PubMedCentralPubMedCrossRef
106.
go back to reference Moldrich RX, Leanage G, She D, Dolan-Evans E, Nelson M, Reza N, et al. Inhibition of histone deacetylase in utero causes sociability deficits in postnatal mice. Behav Brain Res. 2013;257:253–64.PubMedCrossRef Moldrich RX, Leanage G, She D, Dolan-Evans E, Nelson M, Reza N, et al. Inhibition of histone deacetylase in utero causes sociability deficits in postnatal mice. Behav Brain Res. 2013;257:253–64.PubMedCrossRef
107.
go back to reference Chiocchetti AG, Bour HS, Freitag CM. Glutamatergic candidate genes in autism spectrum disorder: an overview. J Neural Transm. 2014;121:1081–106.PubMedCrossRef Chiocchetti AG, Bour HS, Freitag CM. Glutamatergic candidate genes in autism spectrum disorder: an overview. J Neural Transm. 2014;121:1081–106.PubMedCrossRef
108.
go back to reference Rojas DC. The role of glutamate and its receptors in autism and the use of glutamate receptor antagonists in treatment. J Neural Transm. 2014;121:891–905.PubMedCentralPubMedCrossRef Rojas DC. The role of glutamate and its receptors in autism and the use of glutamate receptor antagonists in treatment. J Neural Transm. 2014;121:891–905.PubMedCentralPubMedCrossRef
109.
go back to reference Shimmura C, Suda S, Tsuchiya KJ, Hashimoto K, Ohno K, Matsuzaki H, et al. Alteration of plasma glutamate and glutamine levels in children with high-functioning autism. PLoS One. 2011;6:e25340.PubMedCentralPubMedCrossRef Shimmura C, Suda S, Tsuchiya KJ, Hashimoto K, Ohno K, Matsuzaki H, et al. Alteration of plasma glutamate and glutamine levels in children with high-functioning autism. PLoS One. 2011;6:e25340.PubMedCentralPubMedCrossRef
110.
go back to reference Steeb H, Ramsey JM, Guest PC, Stocki P, Cooper JD, Rahmoune H, et al. Serum proteomic analysis identifies sex-specific differences in lipid metabolism and inflammation profiles in adults diagnosed with Asperger syndrome. Mol Autism. 2014;5:4.PubMedCentralPubMedCrossRef Steeb H, Ramsey JM, Guest PC, Stocki P, Cooper JD, Rahmoune H, et al. Serum proteomic analysis identifies sex-specific differences in lipid metabolism and inflammation profiles in adults diagnosed with Asperger syndrome. Mol Autism. 2014;5:4.PubMedCentralPubMedCrossRef
111.
go back to reference Nuber UA, Kriaucionis S, Roloff TC, Guy J, Selfridge J, Steinhoff C, et al. Up-regulation of glucocorticoid-regulated genes in a mouse model of Rett syndrome. Hum Mol Genet. 2005;14:2247–56.PubMedCrossRef Nuber UA, Kriaucionis S, Roloff TC, Guy J, Selfridge J, Steinhoff C, et al. Up-regulation of glucocorticoid-regulated genes in a mouse model of Rett syndrome. Hum Mol Genet. 2005;14:2247–56.PubMedCrossRef
112.
go back to reference Laurence JA, Fatemi SH. Glial fibrillary acidic protein is elevated in superior frontal, parietal and cerebellar cortices of autistic subjects. Cerebellum. 2005;4:206–10.PubMedCrossRef Laurence JA, Fatemi SH. Glial fibrillary acidic protein is elevated in superior frontal, parietal and cerebellar cortices of autistic subjects. Cerebellum. 2005;4:206–10.PubMedCrossRef
113.
go back to reference Neale BM, Kou Y, Liu L, Ma'ayan A, Samocha KE, Sabo A, et al. Patterns and rates of exonic de novo mutations in autism spectrum disorders. Nature. 2012;485:242–5.PubMedCentralPubMedCrossRef Neale BM, Kou Y, Liu L, Ma'ayan A, Samocha KE, Sabo A, et al. Patterns and rates of exonic de novo mutations in autism spectrum disorders. Nature. 2012;485:242–5.PubMedCentralPubMedCrossRef
114.
go back to reference Main PA, Angley MT, O'Doherty CE, Thomas P, Fenech M. The potential role of the antioxidant and detoxification properties of glutathione in autism spectrum disorders: a systematic review and meta-analysis. Nutr Metab (Lond). 2012;9:35.CrossRef Main PA, Angley MT, O'Doherty CE, Thomas P, Fenech M. The potential role of the antioxidant and detoxification properties of glutathione in autism spectrum disorders: a systematic review and meta-analysis. Nutr Metab (Lond). 2012;9:35.CrossRef
115.
go back to reference Martin J, Cooper M, Hamshere ML, Pocklington A, Scherer SW, Kent L, et al. Biological overlap of attention-deficit/hyperactivity disorder and autism spectrum disorder: evidence from copy number variants. J Am Acad Child Adolesc Psychiatry. 2014;53:761–70. e726.PubMedCentralPubMedCrossRef Martin J, Cooper M, Hamshere ML, Pocklington A, Scherer SW, Kent L, et al. Biological overlap of attention-deficit/hyperactivity disorder and autism spectrum disorder: evidence from copy number variants. J Am Acad Child Adolesc Psychiatry. 2014;53:761–70. e726.PubMedCentralPubMedCrossRef
116.
go back to reference Ramsey JM, Guest PC, Broek JA, Glennon JC, Rommelse N, Franke B, et al. Identification of an age-dependent biomarker signature in children and adolescents with autism spectrum disorders. Mol Autism. 2013;4:27.PubMedCentralPubMedCrossRef Ramsey JM, Guest PC, Broek JA, Glennon JC, Rommelse N, Franke B, et al. Identification of an age-dependent biomarker signature in children and adolescents with autism spectrum disorders. Mol Autism. 2013;4:27.PubMedCentralPubMedCrossRef
117.
go back to reference Molin AM, Andrieux J, Koolen DA, Malan V, Carella M, Colleaux L, et al. A novel microdeletion syndrome at 3q13.31 characterised by developmental delay, postnatal overgrowth, hypoplastic male genitals, and characteristic facial features. J Med Genet. 2012;49:104–9.PubMedCentralPubMedCrossRef Molin AM, Andrieux J, Koolen DA, Malan V, Carella M, Colleaux L, et al. A novel microdeletion syndrome at 3q13.31 characterised by developmental delay, postnatal overgrowth, hypoplastic male genitals, and characteristic facial features. J Med Genet. 2012;49:104–9.PubMedCentralPubMedCrossRef
118.
go back to reference Hu VW, Frank BC, Heine S, Lee NH, Quackenbush J. Gene expression profiling of lymphoblastoid cell lines from monozygotic twins discordant in severity of autism reveals differential regulation of neurologically relevant genes. BMC Genomics. 2006;7:118.PubMedCentralPubMedCrossRef Hu VW, Frank BC, Heine S, Lee NH, Quackenbush J. Gene expression profiling of lymphoblastoid cell lines from monozygotic twins discordant in severity of autism reveals differential regulation of neurologically relevant genes. BMC Genomics. 2006;7:118.PubMedCentralPubMedCrossRef
119.
go back to reference Kenny EM, Cormican P, Furlong S, Heron E, Kenny G, Fahey C, et al. Excess of rare novel loss-of-function variants in synaptic genes in schizophrenia and autism spectrum disorders. Mol Psychiatry. 2014;19:872–9.PubMedCrossRef Kenny EM, Cormican P, Furlong S, Heron E, Kenny G, Fahey C, et al. Excess of rare novel loss-of-function variants in synaptic genes in schizophrenia and autism spectrum disorders. Mol Psychiatry. 2014;19:872–9.PubMedCrossRef
120.
go back to reference Vaags AK, Lionel AC, Sato D, Goodenberger M, Stein QP, Curran S, et al. Rare deletions at the neurexin 3 locus in autism spectrum disorder. Am J Hum Genet. 2012;90:133–41.PubMedCentralPubMedCrossRef Vaags AK, Lionel AC, Sato D, Goodenberger M, Stein QP, Curran S, et al. Rare deletions at the neurexin 3 locus in autism spectrum disorder. Am J Hum Genet. 2012;90:133–41.PubMedCentralPubMedCrossRef
121.
go back to reference Li J, Shi M, Ma Z, Zhao S, Euskirchen G, Ziskin J, et al. Integrated systems analysis reveals a molecular network underlying autism spectrum disorders. Mol Syst Biol. 2014;10:774. Li J, Shi M, Ma Z, Zhao S, Euskirchen G, Ziskin J, et al. Integrated systems analysis reveals a molecular network underlying autism spectrum disorders. Mol Syst Biol. 2014;10:774.
122.
go back to reference Autism Genome Project C, Szatmari P, Paterson AD, Zwaigenbaum L, Roberts W, Brian J, Liu XQ, et al. Mapping autism risk loci using genetic linkage and chromosomal rearrangements. Nat Genet. 2007;39:319–328. Autism Genome Project C, Szatmari P, Paterson AD, Zwaigenbaum L, Roberts W, Brian J, Liu XQ, et al. Mapping autism risk loci using genetic linkage and chromosomal rearrangements. Nat Genet. 2007;39:319–328.
Metadata
Title
Overexpression of Homer1a in the basal and lateral amygdala impairs fear conditioning and induces an autism-like social impairment
Authors
Anwesha Banerjee
Jonathan A. Luong
Anthony Ho
Aeshah O. Saib
Jonathan E. Ploski
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Molecular Autism / Issue 1/2016
Electronic ISSN: 2040-2392
DOI
https://doi.org/10.1186/s13229-016-0077-9

Other articles of this Issue 1/2016

Molecular Autism 1/2016 Go to the issue