Skip to main content
Top
Published in: Molecular Autism 1/2017

Open Access 01-12-2017 | Research

Developmental disruption of amygdala transcriptome and socioemotional behavior in rats exposed to valproic acid prenatally

Authors: Catherine E. Barrett, Thomas M. Hennessey, Katelyn M. Gordon, Steve J. Ryan, Morgan L. McNair, Kerry J. Ressler, Donald G. Rainnie

Published in: Molecular Autism | Issue 1/2017

Login to get access

Abstract

Background

The amygdala controls socioemotional behavior and has consistently been implicated in the etiology of autism spectrum disorder (ASD). Precocious amygdala development is commonly reported in ASD youth with the degree of overgrowth positively correlated to the severity of ASD symptoms. Prenatal exposure to VPA leads to an ASD phenotype in both humans and rats and has become a commonly used tool to model the complexity of ASD symptoms in the laboratory. Here, we examined abnormalities in gene expression in the amygdala and socioemotional behavior across development in the valproic acid (VPA) rat model of ASD.

Methods

Rat dams received oral gavage of VPA (500 mg/kg) or saline daily between E11 and 13. Socioemotional behavior was tracked across development in both sexes. RNA sequencing and proteomics were performed on amygdala samples from male rats across development.

Results

Effects of VPA on time spent in social proximity and anxiety-like behavior were sex dependent, with social abnormalities presenting in males and heightened anxiety in females. Across time VPA stunted developmental and immune, but enhanced cellular death and disorder, pathways in the amygdala relative to saline controls. At postnatal day 10, gene pathways involved in nervous system and cellular development displayed predicted activations in prenatally exposed VPA amygdala samples. By juvenile age, however, transcriptomic and proteomic pathways displayed reductions in cellular growth and neural development. Alterations in immune pathways, calcium signaling, Rho GTPases, and protein kinase A signaling were also observed.

Conclusions

As behavioral, developmental, and genomic alterations are similar to those reported in ASD, these results lend support to prenatal exposure to VPA as a useful tool for understanding how developmental insults to molecular pathways in the amygdala give rise to ASD-related syndromes.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ornoy A, Weinstein-Fudim L, Ergaz Z. Prenatal factors associated with autism spectrum disorder (ASD). Reprod Toxicol. 2015;56:155–69.PubMedCrossRef Ornoy A, Weinstein-Fudim L, Ergaz Z. Prenatal factors associated with autism spectrum disorder (ASD). Reprod Toxicol. 2015;56:155–69.PubMedCrossRef
2.
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(8):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(8):551–5.PubMedCrossRef
3.
go back to reference Christensen J, Gronborg TK, Sorensen MJ, Schendel D, Parner ET, Pedersen LH, et al. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA. 2013;309(16):1696–703.PubMedPubMedCentralCrossRef Christensen J, Gronborg TK, Sorensen MJ, Schendel D, Parner ET, Pedersen LH, et al. Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism. JAMA. 2013;309(16):1696–703.PubMedPubMedCentralCrossRef
4.
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(7):489–97.PubMedPubMedCentralCrossRef 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(7):489–97.PubMedPubMedCentralCrossRef
5.
go back to reference Arndt TL, Stodgell CJ, Rodier PM. The teratology of autism. Int J Dev Neurosci. 2005;23(2-3):189–99.PubMedCrossRef Arndt TL, Stodgell CJ, Rodier PM. The teratology of autism. Int J Dev Neurosci. 2005;23(2-3):189–99.PubMedCrossRef
6.
go back to reference Roullet FI, Lai JK, Foster JA. In utero exposure to valproic acid and autism--a current review of clinical and animal studies. Neurotoxicol Teratol. 2013;36:47–56.PubMedCrossRef Roullet FI, Lai JK, Foster JA. In utero exposure to valproic acid and autism--a current review of clinical and animal studies. Neurotoxicol Teratol. 2013;36:47–56.PubMedCrossRef
7.
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(6):1309–18.PubMedCrossRef 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(6):1309–18.PubMedCrossRef
8.
go back to reference Schneider T, Przewlocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology. 2005;30(1):80–9.PubMedCrossRef Schneider T, Przewlocki R. Behavioral alterations in rats prenatally exposed to valproic acid: animal model of autism. Neuropsychopharmacology. 2005;30(1):80–9.PubMedCrossRef
9.
go back to reference American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th ed. Arlington, VA: American Psychiatric Publishing; 2013.CrossRef American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th ed. Arlington, VA: American Psychiatric Publishing; 2013.CrossRef
10.
go back to reference Baron-Cohen S, Ring HA, Bullmore ET, Wheelwright S, Ashwin C, Williams SC. The amygdala theory of autism. Neurosci Biobehav Rev. 2000;24(3):355–64.PubMedCrossRef Baron-Cohen S, Ring HA, Bullmore ET, Wheelwright S, Ashwin C, Williams SC. The amygdala theory of autism. Neurosci Biobehav Rev. 2000;24(3):355–64.PubMedCrossRef
11.
go back to reference Aylward EH, Minshew NJ, Goldstein G, Honeycutt NA, Augustine AM, Yates KO, et al. MRI volumes of amygdala and hippocampus in non-mentally retarded autistic adolescents and adults. Neurology. 1999;53(9):2145–50.PubMedCrossRef Aylward EH, Minshew NJ, Goldstein G, Honeycutt NA, Augustine AM, Yates KO, et al. MRI volumes of amygdala and hippocampus in non-mentally retarded autistic adolescents and adults. Neurology. 1999;53(9):2145–50.PubMedCrossRef
12.
go back to reference Nacewicz BM, Dalton KM, Johnstone T, Long MT, McAuliff EM, Oakes TR, et al. Amygdala volume and nonverbal social impairment in adolescent and adult males with autism. Arch Gen Psychiatry. 2006;63(12):1417–28.PubMedPubMedCentralCrossRef Nacewicz BM, Dalton KM, Johnstone T, Long MT, McAuliff EM, Oakes TR, et al. Amygdala volume and nonverbal social impairment in adolescent and adult males with autism. Arch Gen Psychiatry. 2006;63(12):1417–28.PubMedPubMedCentralCrossRef
13.
go back to reference Rojas DC, Smith JA, Benkers TL, Camou SL, Reite ML, Rogers SJ. Hippocampus and amygdala volumes in parents of children with autistic disorder. Am J Psychiatry. 2004;161(11):2038–44.PubMedCrossRef Rojas DC, Smith JA, Benkers TL, Camou SL, Reite ML, Rogers SJ. Hippocampus and amygdala volumes in parents of children with autistic disorder. Am J Psychiatry. 2004;161(11):2038–44.PubMedCrossRef
14.
go back to reference Palmen SJ, van Engeland H, Hof PR, Schmitz C. Neuropathological findings in autism. Brain. 2004;127(Pt 12):2572–83.PubMedCrossRef Palmen SJ, van Engeland H, Hof PR, Schmitz C. Neuropathological findings in autism. Brain. 2004;127(Pt 12):2572–83.PubMedCrossRef
15.
go back to reference Schumann CM, Amaral DG. Stereological analysis of amygdala neuron number in autism. J Neurosci. 2006;26(29):7674–9.PubMedCrossRef Schumann CM, Amaral DG. Stereological analysis of amygdala neuron number in autism. J Neurosci. 2006;26(29):7674–9.PubMedCrossRef
16.
go back to reference Dalton KM, Nacewicz BM, Johnstone T, Schaefer HS, Gernsbacher MA, Goldsmith HH, et al. Gaze fixation and the neural circuitry of face processing in autism. Nat Neurosci. 2005;8(4):519–26.PubMedPubMedCentral Dalton KM, Nacewicz BM, Johnstone T, Schaefer HS, Gernsbacher MA, Goldsmith HH, et al. Gaze fixation and the neural circuitry of face processing in autism. Nat Neurosci. 2005;8(4):519–26.PubMedPubMedCentral
17.
go back to reference Hadjikhani N, Joseph RM, Snyder J, Tager-Flusberg H. Abnormal activation of the social brain during face perception in autism. Hum Brain Mapp. 2007;28(5):441–9.PubMedCrossRef Hadjikhani N, Joseph RM, Snyder J, Tager-Flusberg H. Abnormal activation of the social brain during face perception in autism. Hum Brain Mapp. 2007;28(5):441–9.PubMedCrossRef
18.
go back to reference Wang AT, Dapretto M, Hariri AR, Sigman M, Bookheimer SY. Neural correlates of facial affect processing in children and adolescents with autism spectrum disorder. J Am Acad Child Adolesc Psychiatry. 2004;43(4):481–90.PubMedCrossRef Wang AT, Dapretto M, Hariri AR, Sigman M, Bookheimer SY. Neural correlates of facial affect processing in children and adolescents with autism spectrum disorder. J Am Acad Child Adolesc Psychiatry. 2004;43(4):481–90.PubMedCrossRef
19.
go back to reference Baron-Cohen S, Ring HA, Wheelwright S, Bullmore ET, Brammer MJ, Simmons A, et al. Social intelligence in the normal and autistic brain: an fMRI study. Eur J Neurosci. 1999;11(6):1891–8.PubMedCrossRef Baron-Cohen S, Ring HA, Wheelwright S, Bullmore ET, Brammer MJ, Simmons A, et al. Social intelligence in the normal and autistic brain: an fMRI study. Eur J Neurosci. 1999;11(6):1891–8.PubMedCrossRef
20.
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(2-3):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(2-3):125–41.PubMedCrossRef
21.
go back to reference Weng SJ, Carrasco M, Swartz JR, Wiggins JL, Kurapati N, Liberzon I, et al. Neural activation to emotional faces in adolescents with autism spectrum disorders. J Child Psychol Psychiatry. 2011;52(3):296–305.PubMedCrossRef Weng SJ, Carrasco M, Swartz JR, Wiggins JL, Kurapati N, Liberzon I, et al. Neural activation to emotional faces in adolescents with autism spectrum disorders. J Child Psychol Psychiatry. 2011;52(3):296–305.PubMedCrossRef
22.
go back to reference Monk CS, Weng SJ, Wiggins JL, Kurapati N, Louro HM, Carrasco M, et al. Neural circuitry of emotional face processing in autism spectrum disorders. J Psychiatry Neurosci. 2010;35(2):105–14.PubMedPubMedCentralCrossRef Monk CS, Weng SJ, Wiggins JL, Kurapati N, Louro HM, Carrasco M, et al. Neural circuitry of emotional face processing in autism spectrum disorders. J Psychiatry Neurosci. 2010;35(2):105–14.PubMedPubMedCentralCrossRef
23.
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(4):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(4):901–12.PubMedCrossRef
24.
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(1):e55248.PubMedPubMedCentralCrossRef 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(1):e55248.PubMedPubMedCentralCrossRef
25.
go back to reference Bringas ME, Carvajal-Flores FN, Lopez-Ramirez TA, Atzori M, Flores G. Rearrangement of the dendritic morphology in limbic regions and altered exploratory behavior in a rat model of autism spectrum disorder. Neuroscience. 2013;241:170–87.PubMedCrossRef Bringas ME, Carvajal-Flores FN, Lopez-Ramirez TA, Atzori M, Flores G. Rearrangement of the dendritic morphology in limbic regions and altered exploratory behavior in a rat model of autism spectrum disorder. Neuroscience. 2013;241:170–87.PubMedCrossRef
26.
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.PubMedPubMedCentral 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.PubMedPubMedCentral
27.
go back to reference Sui L, Chen M. Prenatal exposure to valproic acid enhances synaptic plasticity in the medial prefrontal cortex and fear memories. Brain Res Bull. 2012;87(6):556–63.PubMedCrossRef Sui L, Chen M. Prenatal exposure to valproic acid enhances synaptic plasticity in the medial prefrontal cortex and fear memories. Brain Res Bull. 2012;87(6):556–63.PubMedCrossRef
29.
go back to reference Rubenstein JL, Merzenich MM. Model of autism: increased ratio of excitation/inhibition in key neural systems. Genes Brain Behav. 2003;2(5):255–67.PubMedCrossRef Rubenstein JL, Merzenich MM. Model of autism: increased ratio of excitation/inhibition in key neural systems. Genes Brain Behav. 2003;2(5):255–67.PubMedCrossRef
30.
go back to reference Rippon G, Brock J, Brown C, Boucher J. Disordered connectivity in the autistic brain: challenges for the “new psychophysiology”. Int J Psychophysiol. 2007;63(2):164–72.PubMedCrossRef Rippon G, Brock J, Brown C, Boucher J. Disordered connectivity in the autistic brain: challenges for the “new psychophysiology”. Int J Psychophysiol. 2007;63(2):164–72.PubMedCrossRef
31.
go back to reference Just MA, Cherkassky VL, Keller TA, Minshew NJ. Cortical activation and synchronization during sentence comprehension in high-functioning autism: evidence of underconnectivity. Brain. 2004;127(Pt 8):1811–21.PubMedCrossRef Just MA, Cherkassky VL, Keller TA, Minshew NJ. Cortical activation and synchronization during sentence comprehension in high-functioning autism: evidence of underconnectivity. Brain. 2004;127(Pt 8):1811–21.PubMedCrossRef
32.
go back to reference Nordahl CW, Scholz R, Yang X, Buonocore MH, Simon T, Rogers S, et al. Increased rate of amygdala growth in children aged 2 to 4 years with autism spectrum disorders: a longitudinal study. Arch Gen Psychiatry. 2012;69(1):53–61.PubMedPubMedCentralCrossRef Nordahl CW, Scholz R, Yang X, Buonocore MH, Simon T, Rogers S, et al. Increased rate of amygdala growth in children aged 2 to 4 years with autism spectrum disorders: a longitudinal study. Arch Gen Psychiatry. 2012;69(1):53–61.PubMedPubMedCentralCrossRef
33.
go back to reference Sparks BF, Friedman SD, Shaw DW, Aylward EH, Echelard D, Artru AA, et al. Brain structural abnormalities in young children with autism spectrum disorder. Neurology. 2002;59(2):184–92.PubMedCrossRef Sparks BF, Friedman SD, Shaw DW, Aylward EH, Echelard D, Artru AA, et al. Brain structural abnormalities in young children with autism spectrum disorder. Neurology. 2002;59(2):184–92.PubMedCrossRef
34.
go back to reference Schumann CM, Hamstra J, Goodlin-Jones BL, Lotspeich LJ, Kwon H, Buonocore MH, et al. The amygdala is enlarged in children but not adolescents with autism; The hippocampus is enlarged at all ages. J Neurosci. 2004;24(28):6392–401.PubMedCrossRef Schumann CM, Hamstra J, Goodlin-Jones BL, Lotspeich LJ, Kwon H, Buonocore MH, et al. The amygdala is enlarged in children but not adolescents with autism; The hippocampus is enlarged at all ages. J Neurosci. 2004;24(28):6392–401.PubMedCrossRef
35.
go back to reference Costello EJ, Egger HL, Angold A. The developmental epidemiology of anxiety disorders: phenomenology, prevalence, and comorbidity. Child Adolesc Psychiatr Clin N Am. 2005;14(4):631–48. vii.PubMedCrossRef Costello EJ, Egger HL, Angold A. The developmental epidemiology of anxiety disorders: phenomenology, prevalence, and comorbidity. Child Adolesc Psychiatr Clin N Am. 2005;14(4):631–48. vii.PubMedCrossRef
36.
go back to reference van Steensel FJ, Bogels SM, Perrin S. Anxiety disorders in children and adolescents with autistic spectrum disorders: a meta-analysis. Clin Child Fam Psychol Rev. 2011;14(3):302–17.PubMedPubMedCentralCrossRef van Steensel FJ, Bogels SM, Perrin S. Anxiety disorders in children and adolescents with autistic spectrum disorders: a meta-analysis. Clin Child Fam Psychol Rev. 2011;14(3):302–17.PubMedPubMedCentralCrossRef
37.
go back to reference White SW, Albano AM, Johnson CR, Kasari C, Ollendick T, Klin A, et al. Development of a cognitive-behavioral intervention program to treat anxiety and social deficits in teens with high-functioning autism. Clin Child Fam Psychol Rev. 2010;13(1):77–90.PubMedPubMedCentralCrossRef White SW, Albano AM, Johnson CR, Kasari C, Ollendick T, Klin A, et al. Development of a cognitive-behavioral intervention program to treat anxiety and social deficits in teens with high-functioning autism. Clin Child Fam Psychol Rev. 2010;13(1):77–90.PubMedPubMedCentralCrossRef
39.
go back to reference Ehrlich DE, Ryan SJ, Hazra R, Guo JD, Rainnie DG. Postnatal maturation of GABAergic transmission in the rat basolateral amygdala. J Neurophysiol. 2013;110(4):926–41.PubMedPubMedCentralCrossRef Ehrlich DE, Ryan SJ, Hazra R, Guo JD, Rainnie DG. Postnatal maturation of GABAergic transmission in the rat basolateral amygdala. J Neurophysiol. 2013;110(4):926–41.PubMedPubMedCentralCrossRef
40.
go back to reference Ehrlich DE, Ryan SJ, Rainnie DG. Postnatal development of electrophysiological properties of principal neurons in the rat basolateral amygdala. J Physiol. 2012;590(19):4819–38.PubMedPubMedCentralCrossRef Ehrlich DE, Ryan SJ, Rainnie DG. Postnatal development of electrophysiological properties of principal neurons in the rat basolateral amygdala. J Physiol. 2012;590(19):4819–38.PubMedPubMedCentralCrossRef
41.
go back to reference Ehrlich DE, Ryan SJ, Rainnie DG. Postnatal development of electrophysiological properties of principal neurons in the rat basolateral amygdala. J Physiol (Lond). 2012;590(19):4819–38.CrossRef Ehrlich DE, Ryan SJ, Rainnie DG. Postnatal development of electrophysiological properties of principal neurons in the rat basolateral amygdala. J Physiol (Lond). 2012;590(19):4819–38.CrossRef
42.
go back to reference Ryan SJ, Ehrlich DE, Rainnie DG. Morphology and dendritic maturation of developing principal neurons in the rat basolateral amygdala. Brain Struct Funct. 2016;221(2):839–54.PubMedCrossRef Ryan SJ, Ehrlich DE, Rainnie DG. Morphology and dendritic maturation of developing principal neurons in the rat basolateral amygdala. Brain Struct Funct. 2016;221(2):839–54.PubMedCrossRef
43.
go back to reference Sullivan RM, Landers M, Yeaman B, Wilson DA. Good memories of bad events in infancy. Nature. 2000;407(6800):38–9.PubMedCrossRef Sullivan RM, Landers M, Yeaman B, Wilson DA. Good memories of bad events in infancy. Nature. 2000;407(6800):38–9.PubMedCrossRef
44.
go back to reference Thompson JV, Sullivan RM, Wilson DA. Developmental emergence of fear learning corresponds with changes in amygdala synaptic plasticity. Brain Res. 2008;1200:58–65.PubMedPubMedCentralCrossRef Thompson JV, Sullivan RM, Wilson DA. Developmental emergence of fear learning corresponds with changes in amygdala synaptic plasticity. Brain Res. 2008;1200:58–65.PubMedPubMedCentralCrossRef
45.
go back to reference Brummelte S, Witte V, Teuchert-Noodt G. Postnatal development of GABA and calbindin cells and fibers in the prefrontal cortex and basolateral amygdala of gerbils (Meriones unguiculatus). Int J Dev Neurosci. 2007;25(3):191–200.PubMedCrossRef Brummelte S, Witte V, Teuchert-Noodt G. Postnatal development of GABA and calbindin cells and fibers in the prefrontal cortex and basolateral amygdala of gerbils (Meriones unguiculatus). Int J Dev Neurosci. 2007;25(3):191–200.PubMedCrossRef
46.
go back to reference Kim JH, McNally GP, Richardson R. Recovery of fear memories in rats: Role of gamma-amino butyric acid (GABA) in infantile amnesia. Behav Neurosci. 2006;120(1):40–8.PubMedCrossRef Kim JH, McNally GP, Richardson R. Recovery of fear memories in rats: Role of gamma-amino butyric acid (GABA) in infantile amnesia. Behav Neurosci. 2006;120(1):40–8.PubMedCrossRef
47.
go back to reference Li C, Rainnie DG. Bidirectional regulation of synaptic plasticity in the basolateral amygdala induced by the D1-like family of dopamine receptors and group II metabotropic glutamate receptors. J Physiol. 2014;592(19):4329–51.PubMedPubMedCentralCrossRef Li C, Rainnie DG. Bidirectional regulation of synaptic plasticity in the basolateral amygdala induced by the D1-like family of dopamine receptors and group II metabotropic glutamate receptors. J Physiol. 2014;592(19):4329–51.PubMedPubMedCentralCrossRef
48.
go back to reference Ryan SJ, Ehrlich DE, Jasnow AM, Daftary S, Madsen TE, Rainnie DG. Spike-timing precision and neuronal synchrony are enhanced by an interaction between synaptic inhibition and membrane oscillations in the amygdala. PLoS One. 2012;7(4):e35320.PubMedPubMedCentralCrossRef Ryan SJ, Ehrlich DE, Jasnow AM, Daftary S, Madsen TE, Rainnie DG. Spike-timing precision and neuronal synchrony are enhanced by an interaction between synaptic inhibition and membrane oscillations in the amygdala. PLoS One. 2012;7(4):e35320.PubMedPubMedCentralCrossRef
49.
go back to reference Favre MR, Barkat TR, Lamendola D, Khazen G, Markram H, Markram K. General developmental health in the VPA-rat model of autism. Front Behav Neurosci. 2013;7:88.PubMedPubMedCentralCrossRef Favre MR, Barkat TR, Lamendola D, Khazen G, Markram H, Markram K. General developmental health in the VPA-rat model of autism. Front Behav Neurosci. 2013;7:88.PubMedPubMedCentralCrossRef
50.
go back to reference Nau H. Transfer of valproic acid and its main active unsaturated metabolite to the gestational tissue: correlation with neural tube defect formation in the mouse. Teratology. 1986;33(1):21–7.PubMedCrossRef Nau H. Transfer of valproic acid and its main active unsaturated metabolite to the gestational tissue: correlation with neural tube defect formation in the mouse. Teratology. 1986;33(1):21–7.PubMedCrossRef
51.
go back to reference Todrank J, Heth G, Restrepo D. Effects of in utero odorant exposure on neuroanatomical development of the olfactory bulb and odour preferences. Proc Biol Sci. 2011;278(1714):1949–55.PubMedCrossRef Todrank J, Heth G, Restrepo D. Effects of in utero odorant exposure on neuroanatomical development of the olfactory bulb and odour preferences. Proc Biol Sci. 2011;278(1714):1949–55.PubMedCrossRef
52.
go back to reference Dias BG, Ressler KJ. Parental olfactory experience influences behavior and neural structure in subsequent generations. Nat Neurosci. 2014;17(1):89–96.PubMedCrossRef Dias BG, Ressler KJ. Parental olfactory experience influences behavior and neural structure in subsequent generations. Nat Neurosci. 2014;17(1):89–96.PubMedCrossRef
53.
go back to reference Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21.PubMedCrossRef Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15–21.PubMedCrossRef
54.
go back to reference Anders S, Pyl PT, Huber W. HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31(2):166–9.PubMedCrossRef Anders S, Pyl PT, Huber W. HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31(2):166–9.PubMedCrossRef
56.
go back to reference Kramer A, Green J, Pollard Jr J, Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014;30(4):523–30.PubMedCrossRef Kramer A, Green J, Pollard Jr J, Tugendreich S. Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics. 2014;30(4):523–30.PubMedCrossRef
57.
go back to reference Schneider T, Roman A, Basta-Kaim A, Kubera M, Budziszewska B, Schneider K, et al. Gender-specific behavioral and immunological alterations in an animal model of autism induced by prenatal exposure to valproic acid. Psychoneuroendocrinology. 2008;33(6):728–40.PubMedCrossRef Schneider T, Roman A, Basta-Kaim A, Kubera M, Budziszewska B, Schneider K, et al. Gender-specific behavioral and immunological alterations in an animal model of autism induced by prenatal exposure to valproic acid. Psychoneuroendocrinology. 2008;33(6):728–40.PubMedCrossRef
58.
go back to reference Schneider T, Turczak J, Przewlocki R. Environmental enrichment reverses behavioral alterations in rats prenatally exposed to valproic acid: issues for a therapeutic approach in autism. Neuropsychopharmacology. 2006;31(1):36–46.PubMed Schneider T, Turczak J, Przewlocki R. Environmental enrichment reverses behavioral alterations in rats prenatally exposed to valproic acid: issues for a therapeutic approach in autism. Neuropsychopharmacology. 2006;31(1):36–46.PubMed
59.
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(2):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(2):514–22.PubMedCrossRef
60.
go back to reference Kim KC, Kim P, Go HS, Choi CS, Park JH, Kim HJ, et al. Male-specific alteration in excitatory post-synaptic development and social interaction in pre-natal valproic acid exposure model of autism spectrum disorder. J Neurochem. 2013;124(6):832–43.PubMedCrossRef Kim KC, Kim P, Go HS, Choi CS, Park JH, Kim HJ, et al. Male-specific alteration in excitatory post-synaptic development and social interaction in pre-natal valproic acid exposure model of autism spectrum disorder. J Neurochem. 2013;124(6):832–43.PubMedCrossRef
61.
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
62.
go back to reference Grillon C. Startle reactivity and anxiety disorders: aversive conditioning, context, and neurobiology. Biol Psychiatry. 2002;52(10):958–75.PubMedCrossRef Grillon C. Startle reactivity and anxiety disorders: aversive conditioning, context, and neurobiology. Biol Psychiatry. 2002;52(10):958–75.PubMedCrossRef
63.
go back to reference Chamberlain PD, Rodgers J, Crowley MJ, White SE, Freeston MH, South M. A potentiated startle study of uncertainty and contextual anxiety in adolescents diagnosed with autism spectrum disorder. Mol Autism. 2013;4(1):31.PubMedPubMedCentralCrossRef Chamberlain PD, Rodgers J, Crowley MJ, White SE, Freeston MH, South M. A potentiated startle study of uncertainty and contextual anxiety in adolescents diagnosed with autism spectrum disorder. Mol Autism. 2013;4(1):31.PubMedPubMedCentralCrossRef
64.
go back to reference Christensen DL, Baio J, Van Naarden BK, Bilder D, Charles J, Constantino JN, et al. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years--Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2012. MMWR Surveill Summ. 2016;65(3):1–23.PubMedCrossRef Christensen DL, Baio J, Van Naarden BK, Bilder D, Charles J, Constantino JN, et al. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years--Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2012. MMWR Surveill Summ. 2016;65(3):1–23.PubMedCrossRef
65.
go back to reference Bromley RL, Mawer GE, Briggs M, Cheyne C, Clayton-Smith J, Garcia-Finana M, et al. The prevalence of neurodevelopmental disorders in children prenatally exposed to antiepileptic drugs. J Neurol Neurosurg Psychiatry. 2013;84(6):637–43.PubMedPubMedCentralCrossRef Bromley RL, Mawer GE, Briggs M, Cheyne C, Clayton-Smith J, Garcia-Finana M, et al. The prevalence of neurodevelopmental disorders in children prenatally exposed to antiepileptic drugs. J Neurol Neurosurg Psychiatry. 2013;84(6):637–43.PubMedPubMedCentralCrossRef
66.
go back to reference Kataoka S, Takuma K, Hara Y, Maeda Y, Ago Y, Matsuda T. Autism-like behaviours with transient histone hyperacetylation in mice treated prenatally with valproic acid. Int J Neuropsychopharmacol. 2013;16(1):91–103.PubMedCrossRef Kataoka S, Takuma K, Hara Y, Maeda Y, Ago Y, Matsuda T. Autism-like behaviours with transient histone hyperacetylation in mice treated prenatally with valproic acid. Int J Neuropsychopharmacol. 2013;16(1):91–103.PubMedCrossRef
67.
go back to reference Raza S, Harker A, Richards S, Kolb B, Gibb R. Tactile stimulation improves neuroanatomical pathology but not behavior in rats prenatally exposed to valproic acid. Behav Brain Res. 2015;282:25–36.PubMedCrossRef Raza S, Harker A, Richards S, Kolb B, Gibb R. Tactile stimulation improves neuroanatomical pathology but not behavior in rats prenatally exposed to valproic acid. Behav Brain Res. 2015;282:25–36.PubMedCrossRef
68.
go back to reference Stefanik P, Olexova L, Krskova L. Increased sociability and gene expression of oxytocin and its receptor in the brains of rats affected prenatally by valproic acid. Pharmacol Biochem Behav. 2015;131:42–50.PubMedCrossRef Stefanik P, Olexova L, Krskova L. Increased sociability and gene expression of oxytocin and its receptor in the brains of rats affected prenatally by valproic acid. Pharmacol Biochem Behav. 2015;131:42–50.PubMedCrossRef
69.
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(8):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(8):740–50.PubMedCrossRef
70.
go back to reference Dufour-Rainfray D, Vourc'h P, Le Guisquet AM, Garreau L, Ternant D, Bodard S, et al. Behavior and serotonergic disorders in rats exposed prenatally to valproate: a model for autism. Neurosci Lett. 2010;470(1):55–9.PubMedCrossRef Dufour-Rainfray D, Vourc'h P, Le Guisquet AM, Garreau L, Ternant D, Bodard S, et al. Behavior and serotonergic disorders in rats exposed prenatally to valproate: a model for autism. Neurosci Lett. 2010;470(1):55–9.PubMedCrossRef
71.
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(4):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(4):547–55.CrossRef
72.
go back to reference Nakasato A, Nakatani Y, Seki Y, Tsujino N, Umino M, Arita H. Swim stress exaggerates the hyperactive mesocortical dopamine system in a rodent model of autism. Brain Res. 2008;1193:128–35.PubMedCrossRef Nakasato A, Nakatani Y, Seki Y, Tsujino N, Umino M, Arita H. Swim stress exaggerates the hyperactive mesocortical dopamine system in a rodent model of autism. Brain Res. 2008;1193:128–35.PubMedCrossRef
73.
go back to reference Callaghan BL, Richardson R. The effect of adverse rearing environments on persistent memories in young rats: removing the brakes on infant fear memories. Transl Psychiatry. 2012;2:e138.PubMedPubMedCentralCrossRef Callaghan BL, Richardson R. The effect of adverse rearing environments on persistent memories in young rats: removing the brakes on infant fear memories. Transl Psychiatry. 2012;2:e138.PubMedPubMedCentralCrossRef
75.
76.
go back to reference Scattoni ML, McFarlane HG, Zhodzishsky V, Caldwell HK, Young WS, Ricceri L, et al. Reduced ultrasonic vocalizations in vasopressin 1b knockout mice. Behav Brain Res. 2008;187(2):371–8.PubMedCrossRef Scattoni ML, McFarlane HG, Zhodzishsky V, Caldwell HK, Young WS, Ricceri L, et al. Reduced ultrasonic vocalizations in vasopressin 1b knockout mice. Behav Brain Res. 2008;187(2):371–8.PubMedCrossRef
77.
go back to reference Hofer MA, Brunelli SA, Shair HN. Potentiation of Isolation-Induced Vocalization by Brief Exposure of Rat Pups to Maternal Cues. Dev Psychobiol. 1994;27(8):503–17.PubMedCrossRef Hofer MA, Brunelli SA, Shair HN. Potentiation of Isolation-Induced Vocalization by Brief Exposure of Rat Pups to Maternal Cues. Dev Psychobiol. 1994;27(8):503–17.PubMedCrossRef
78.
go back to reference Hodgson RA, Guthrie DH, Varty GB. Duration of ultrasonic vocalizations in the isolated rat pup as a behavioral measure: Sensitivity to anxiolytic and antidepressant drugs. Pharmacol Biochem Be. 2008;88(3):341–8.CrossRef Hodgson RA, Guthrie DH, Varty GB. Duration of ultrasonic vocalizations in the isolated rat pup as a behavioral measure: Sensitivity to anxiolytic and antidepressant drugs. Pharmacol Biochem Be. 2008;88(3):341–8.CrossRef
79.
go back to reference Esposito G, Del Carmen Rostagno M, Venuti P, Haltigan JD, Messinger DS. Brief Report: Atypical expression of distress during the separation phase of the strange situation procedure in infant siblings at high risk for ASD. J Autism Dev Disord. 2014;44(4):975–80.PubMedPubMedCentralCrossRef Esposito G, Del Carmen Rostagno M, Venuti P, Haltigan JD, Messinger DS. Brief Report: Atypical expression of distress during the separation phase of the strange situation procedure in infant siblings at high risk for ASD. J Autism Dev Disord. 2014;44(4):975–80.PubMedPubMedCentralCrossRef
80.
go back to reference Furlow F. Human neonatal cry quality as an honest signal of fitness. Evol Hum Behav. 1997;18:175–83.CrossRef Furlow F. Human neonatal cry quality as an honest signal of fitness. Evol Hum Behav. 1997;18:175–83.CrossRef
81.
go back to reference Wilson DA, Sullivan RM. Neurobiology of associative learning in the neonate: early olfactory learning. Behav Neural Biol. 1994;61(1):1–18.PubMedCrossRef Wilson DA, Sullivan RM. Neurobiology of associative learning in the neonate: early olfactory learning. Behav Neural Biol. 1994;61(1):1–18.PubMedCrossRef
82.
go back to reference Raineki C, Sarro E, Rincon-Cortes M, Perry R, Boggs J, Holman CJ, et al. Paradoxical neurobehavioral rescue by memories of early-life abuse: the safety signal value of odors learned during abusive attachment. Neuropsychopharmacology. 2015;40(4):906–14.PubMedCrossRef Raineki C, Sarro E, Rincon-Cortes M, Perry R, Boggs J, Holman CJ, et al. Paradoxical neurobehavioral rescue by memories of early-life abuse: the safety signal value of odors learned during abusive attachment. Neuropsychopharmacology. 2015;40(4):906–14.PubMedCrossRef
83.
go back to reference Sevelinges Y, Moriceau S, Holman P, Miner C, Muzny K, Gervais R, et al. Enduring effects of infant memories: infant odor-shock conditioning attenuates amygdala activity and adult fear conditioning. Biol Psychiatry. 2007;62(10):1070–9.PubMedCrossRef Sevelinges Y, Moriceau S, Holman P, Miner C, Muzny K, Gervais R, et al. Enduring effects of infant memories: infant odor-shock conditioning attenuates amygdala activity and adult fear conditioning. Biol Psychiatry. 2007;62(10):1070–9.PubMedCrossRef
84.
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(3):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(3):391–402.PubMedCrossRef
85.
go back to reference Voineagu I, Wang X, Johnston P, Lowe JK, Tian Y, Horvath S, et al. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature. 2011;474(7351):380–4.PubMedPubMedCentralCrossRef Voineagu I, Wang X, Johnston P, Lowe JK, Tian Y, Horvath S, et al. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature. 2011;474(7351):380–4.PubMedPubMedCentralCrossRef
86.
go back to reference Tebbenkamp AT, Willsey AJ, State MW, Sestan N. The developmental transcriptome of the human brain: implications for neurodevelopmental disorders. Curr Opin Neurol. 2014;27(2):149–56.PubMedPubMedCentralCrossRef Tebbenkamp AT, Willsey AJ, State MW, Sestan N. The developmental transcriptome of the human brain: implications for neurodevelopmental disorders. Curr Opin Neurol. 2014;27(2):149–56.PubMedPubMedCentralCrossRef
87.
go back to reference Chow ML, Pramparo T, Winn ME, Barnes CC, Li HR, Weiss L, et al. Age-dependent brain gene expression and copy number anomalies in autism suggest distinct pathological processes at young versus mature ages. PLoS Genet. 2012;8(3):e1002592.PubMedPubMedCentralCrossRef Chow ML, Pramparo T, Winn ME, Barnes CC, Li HR, Weiss L, et al. Age-dependent brain gene expression and copy number anomalies in autism suggest distinct pathological processes at young versus mature ages. PLoS Genet. 2012;8(3):e1002592.PubMedPubMedCentralCrossRef
88.
go back to reference Birnbaum R, Jaffe AE, Hyde TM, Kleinman JE, Weinberger DR. Prenatal expression patterns of genes associated with neuropsychiatric disorders. Am J Psychiatry. 2014;171(7):758–67.PubMedPubMedCentralCrossRef Birnbaum R, Jaffe AE, Hyde TM, Kleinman JE, Weinberger DR. Prenatal expression patterns of genes associated with neuropsychiatric disorders. Am J Psychiatry. 2014;171(7):758–67.PubMedPubMedCentralCrossRef
89.
go back to reference Willsey AJ, Sanders SJ, Li M, Dong S, Tebbenkamp AT, Muhle RA, et al. Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism. Cell. 2013;155(5):997–1007.PubMedPubMedCentralCrossRef Willsey AJ, Sanders SJ, Li M, Dong S, Tebbenkamp AT, Muhle RA, et al. Coexpression networks implicate human midfetal deep cortical projection neurons in the pathogenesis of autism. Cell. 2013;155(5):997–1007.PubMedPubMedCentralCrossRef
90.
go back to reference Parikshak NN, Luo R, Zhang A, Won H, Lowe JK, Chandran V, et al. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell. 2013;155(5):1008–21.PubMedPubMedCentralCrossRef Parikshak NN, Luo R, Zhang A, Won H, Lowe JK, Chandran V, et al. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell. 2013;155(5):1008–21.PubMedPubMedCentralCrossRef
91.
go back to reference Romijn HJ, Hofman MA, Gramsbergen A. At what age is the developing cerebral cortex of the rat comparable to that of the full-term newborn human baby? Early Hum Dev. 1991;26(1):61–7.PubMedCrossRef Romijn HJ, Hofman MA, Gramsbergen A. At what age is the developing cerebral cortex of the rat comparable to that of the full-term newborn human baby? Early Hum Dev. 1991;26(1):61–7.PubMedCrossRef
92.
go back to reference Bakken TE, Miller JA, Ding SL, Sunkin SM, Smith KA, Ng L, et al. A comprehensive transcriptional map of primate brain development. Nature. 2016;535(7612):367–75.PubMedPubMedCentralCrossRef Bakken TE, Miller JA, Ding SL, Sunkin SM, Smith KA, Ng L, et al. A comprehensive transcriptional map of primate brain development. Nature. 2016;535(7612):367–75.PubMedPubMedCentralCrossRef
93.
go back to reference Hao Y, Creson T, Zhang L, Li P, Du F, Yuan P, et al. Mood stabilizer valproate promotes ERK pathway-dependent cortical neuronal growth and neurogenesis. J Neurosci. 2004;24(29):6590–9.PubMedCrossRef Hao Y, Creson T, Zhang L, Li P, Du F, Yuan P, et al. Mood stabilizer valproate promotes ERK pathway-dependent cortical neuronal growth and neurogenesis. J Neurosci. 2004;24(29):6590–9.PubMedCrossRef
94.
go back to reference Wurzman R, Forcelli PA, Griffey CJ, Kromer LF. Repetitive grooming and sensorimotor abnormalities in an ephrin-A knockout model for Autism Spectrum Disorders. Behav Brain Res. 2015;278:115–28.PubMedCrossRef Wurzman R, Forcelli PA, Griffey CJ, Kromer LF. Repetitive grooming and sensorimotor abnormalities in an ephrin-A knockout model for Autism Spectrum Disorders. Behav Brain Res. 2015;278:115–28.PubMedCrossRef
95.
go back to reference Abdallah MW, Larsen N, Mortensen EL, Atladottir HO, Norgaard-Pedersen B, Bonefeld-Jorgensen EC, et al. Neonatal levels of cytokines and risk of autism spectrum disorders: an exploratory register-based historic birth cohort study utilizing the Danish Newborn Screening Biobank. J Neuroimmunol. 2012;252(1-2):75–82.PubMedCrossRef Abdallah MW, Larsen N, Mortensen EL, Atladottir HO, Norgaard-Pedersen B, Bonefeld-Jorgensen EC, et al. Neonatal levels of cytokines and risk of autism spectrum disorders: an exploratory register-based historic birth cohort study utilizing the Danish Newborn Screening Biobank. J Neuroimmunol. 2012;252(1-2):75–82.PubMedCrossRef
96.
go back to reference Estes ML, McAllister AK. Immune mediators in the brain and peripheral tissues in autism spectrum disorder. Nat Rev Neurosci. 2015;16(8):469–86.PubMedCrossRef Estes ML, McAllister AK. Immune mediators in the brain and peripheral tissues in autism spectrum disorder. Nat Rev Neurosci. 2015;16(8):469–86.PubMedCrossRef
97.
go back to reference Kuddo T, Nelson KB. How common are gastrointestinal disorders in children with autism? Curr Opin Pediatr. 2003;15(3):339–43.PubMedCrossRef Kuddo T, Nelson KB. How common are gastrointestinal disorders in children with autism? Curr Opin Pediatr. 2003;15(3):339–43.PubMedCrossRef
98.
go back to reference Enstrom AM, Onore CE, Van de Water JA, Ashwood P. Differential monocyte responses to TLR ligands in children with autism spectrum disorders. Brain Behav Immun. 2010;24(1):64–71.PubMedCrossRef Enstrom AM, Onore CE, Van de Water JA, Ashwood P. Differential monocyte responses to TLR ligands in children with autism spectrum disorders. Brain Behav Immun. 2010;24(1):64–71.PubMedCrossRef
99.
go back to reference Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah I, Van de Water J. Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav Immun. 2011;25(1):40–5.PubMedCrossRef Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah I, Van de Water J. Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav Immun. 2011;25(1):40–5.PubMedCrossRef
100.
go back to reference Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah IN, Van de Water J. Associations of impaired behaviors with elevated plasma chemokines in autism spectrum disorders. J Neuroimmunol. 2011;232(1-2):196–9.PubMedCrossRef Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah IN, Van de Water J. Associations of impaired behaviors with elevated plasma chemokines in autism spectrum disorders. J Neuroimmunol. 2011;232(1-2):196–9.PubMedCrossRef
101.
go back to reference Goines PE, Ashwood P. Cytokine dysregulation in autism spectrum disorders (ASD): possible role of the environment. Neurotoxicol Teratol. 2013;36:67–81.PubMedCrossRef Goines PE, Ashwood P. Cytokine dysregulation in autism spectrum disorders (ASD): possible role of the environment. Neurotoxicol Teratol. 2013;36:67–81.PubMedCrossRef
102.
go back to reference Soueid J, Kourtian S, Makhoul NJ, Makoukji J, Haddad S, Ghanem SS, et al. RYR2, PTDSS1 and AREG genes are implicated in a Lebanese population-based study of copy number variation in autism. Sci Rep. 2016;6:19088.PubMedPubMedCentralCrossRef Soueid J, Kourtian S, Makhoul NJ, Makoukji J, Haddad S, Ghanem SS, et al. RYR2, PTDSS1 and AREG genes are implicated in a Lebanese population-based study of copy number variation in autism. Sci Rep. 2016;6:19088.PubMedPubMedCentralCrossRef
103.
go back to reference Lu AT, Cantor RM. Allowing for sex differences increases power in a GWAS of multiplex Autism families. Mol Psychiatry. 2012;17(2):215–22.PubMedCrossRef Lu AT, Cantor RM. Allowing for sex differences increases power in a GWAS of multiplex Autism families. Mol Psychiatry. 2012;17(2):215–22.PubMedCrossRef
104.
go back to reference Hoppa MB, Lana B, Margas W, Dolphin AC, Ryan TA. alpha2delta expression sets presynaptic calcium channel abundance and release probability. Nature. 2012;486(7401):122–5.PubMedPubMedCentral Hoppa MB, Lana B, Margas W, Dolphin AC, Ryan TA. alpha2delta expression sets presynaptic calcium channel abundance and release probability. Nature. 2012;486(7401):122–5.PubMedPubMedCentral
105.
go back to reference Krey JF, Dolmetsch RE. Molecular mechanisms of autism: a possible role for Ca2+ signaling. Curr Opin Neurobiol. 2007;17(1):112–9.PubMedCrossRef Krey JF, Dolmetsch RE. Molecular mechanisms of autism: a possible role for Ca2+ signaling. Curr Opin Neurobiol. 2007;17(1):112–9.PubMedCrossRef
106.
go back to reference Kitagishi Y, Minami A, Nakanishi A, Ogura Y, Matsuda S. Neuron membrane trafficking and protein kinases involved in autism and ADHD. Int J Mol Sci. 2015;16(2):3095–115.PubMedPubMedCentralCrossRef Kitagishi Y, Minami A, Nakanishi A, Ogura Y, Matsuda S. Neuron membrane trafficking and protein kinases involved in autism and ADHD. Int J Mol Sci. 2015;16(2):3095–115.PubMedPubMedCentralCrossRef
107.
go back to reference Ji L, Chauhan V, Flory MJ, Chauhan A. Brain region-specific decrease in the activity and expression of protein kinase A in the frontal cortex of regressive autism. PLoS One. 2011;6(8):e23751.PubMedPubMedCentralCrossRef Ji L, Chauhan V, Flory MJ, Chauhan A. Brain region-specific decrease in the activity and expression of protein kinase A in the frontal cortex of regressive autism. PLoS One. 2011;6(8):e23751.PubMedPubMedCentralCrossRef
108.
go back to reference Li C, Dabrowska J, Hazra R, Rainnie DG. Synergistic activation of dopamine D1 and TrkB receptors mediate gain control of synaptic plasticity in the basolateral amygdala. PLoS One. 2011;6(10):e26065.PubMedPubMedCentralCrossRef Li C, Dabrowska J, Hazra R, Rainnie DG. Synergistic activation of dopamine D1 and TrkB receptors mediate gain control of synaptic plasticity in the basolateral amygdala. PLoS One. 2011;6(10):e26065.PubMedPubMedCentralCrossRef
109.
go back to reference Lin YC, Frei JA, Kilander MB, Shen W, Blatt GJ. A Subset of Autism-Associated Genes Regulate the Structural Stability of Neurons. Front Cell Neurosci. 2016;10:263.PubMedPubMedCentral Lin YC, Frei JA, Kilander MB, Shen W, Blatt GJ. A Subset of Autism-Associated Genes Regulate the Structural Stability of Neurons. Front Cell Neurosci. 2016;10:263.PubMedPubMedCentral
111.
go back to reference Shapiro LP, Parsons RG, Koleske AJ, Gourley SL. Differential expression of cytoskeletal regulatory factors in the adolescent prefrontal cortex: Implications for cortical development. J Neurosci Res. 2017;95(5):1123–43.PubMedCrossRef Shapiro LP, Parsons RG, Koleske AJ, Gourley SL. Differential expression of cytoskeletal regulatory factors in the adolescent prefrontal cortex: Implications for cortical development. J Neurosci Res. 2017;95(5):1123–43.PubMedCrossRef
112.
go back to reference Zhou Z, Meng Y, Asrar S, Todorovski Z, Jia Z. A critical role of Rho-kinase ROCK2 in the regulation of spine and synaptic function. Neuropharmacology. 2009;56(1):81–9.PubMedCrossRef Zhou Z, Meng Y, Asrar S, Todorovski Z, Jia Z. A critical role of Rho-kinase ROCK2 in the regulation of spine and synaptic function. Neuropharmacology. 2009;56(1):81–9.PubMedCrossRef
113.
go back to reference Vogel C, Marcotte EM. Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet. 2012;13(4):227–32.PubMedPubMedCentral Vogel C, Marcotte EM. Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet. 2012;13(4):227–32.PubMedPubMedCentral
114.
go back to reference Ghazalpour A, Bennett B, Petyuk VA, Orozco L, Hagopian R, Mungrue IN, et al. Comparative analysis of proteome and transcriptome variation in mouse. PLoS Genet. 2011;7(6):e1001393.PubMedPubMedCentralCrossRef Ghazalpour A, Bennett B, Petyuk VA, Orozco L, Hagopian R, Mungrue IN, et al. Comparative analysis of proteome and transcriptome variation in mouse. PLoS Genet. 2011;7(6):e1001393.PubMedPubMedCentralCrossRef
115.
go back to reference Seyfried NT, Dammer EB, Swarup V, Nandakumar D, Duong DM, Yin L, et al. A Multi-network Approach Identifies Protein-Specific Co-expression in Asymptomatic and Symptomatic Alzheimer's Disease. Cell Syst. 2017;4(1):60–72. e4.PubMedCrossRef Seyfried NT, Dammer EB, Swarup V, Nandakumar D, Duong DM, Yin L, et al. A Multi-network Approach Identifies Protein-Specific Co-expression in Asymptomatic and Symptomatic Alzheimer's Disease. Cell Syst. 2017;4(1):60–72. e4.PubMedCrossRef
116.
go back to reference Sharma K, Schmitt S, Bergner CG, Tyanova S, Kannaiyan N, Manrique-Hoyos N, et al. Cell type- and brain region-resolved mouse brain proteome. Nat Neurosci. 2015;18(12):1819–31.PubMedCrossRef Sharma K, Schmitt S, Bergner CG, Tyanova S, Kannaiyan N, Manrique-Hoyos N, et al. Cell type- and brain region-resolved mouse brain proteome. Nat Neurosci. 2015;18(12):1819–31.PubMedCrossRef
117.
go back to reference Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30(1):207–10.PubMedPubMedCentralCrossRef Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30(1):207–10.PubMedPubMedCentralCrossRef
Metadata
Title
Developmental disruption of amygdala transcriptome and socioemotional behavior in rats exposed to valproic acid prenatally
Authors
Catherine E. Barrett
Thomas M. Hennessey
Katelyn M. Gordon
Steve J. Ryan
Morgan L. McNair
Kerry J. Ressler
Donald G. Rainnie
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Molecular Autism / Issue 1/2017
Electronic ISSN: 2040-2392
DOI
https://doi.org/10.1186/s13229-017-0160-x

Other articles of this Issue 1/2017

Molecular Autism 1/2017 Go to the issue