全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

What We Have Learned about Autism Spectrum Disorder from Valproic Acid

DOI: 10.1155/2013/712758

Full-Text   Cite this paper   Add to My Lib

Abstract:

Two recent epidemiological investigations in children exposed to valproic acid (VPA) treatment in utero have reported a significant risk associated with neurodevelopmental disorders and autism spectrum disorder (ASD) in particular. Parallel to this work, there is a growing body of animal research literature using VPA as an animal model of ASD. In this focused review we first summarize the epidemiological evidence linking VPA to ASD and then comment on two important neurobiological findings linking VPA to ASD clinicopathology, namely, accelerated or early brain overgrowth and hyperexcitable networks. Improving our understanding of how the drug VPA can alter early development of neurological systems will ultimately improve our understanding of ASD. 1. Introduction The core clinicopathology of autism spectrum disorder (ASD) is currently thought to be characterized by accelerated or early brain tissue overgrowth in regions involved in emotional, social, and communication functions [1–6]. Although much of the earlier evidence regarding accelerated or early brain overgrowth was based on head circumference measurements which may not be as robust as previously thought [7] (but see [8]), more recent longitudinal brain imaging studies have provided further evidence of this abnormal pattern of brain growth in ASD [3, 6]. In fact, abnormal pathological overgrowth in ASD may even persist into adulthood in certain brain regions [9]. Exposure to exogenous chemicals during pregnancy can interfere with cortical development and the maturation of offspring. One such exogenous chemical is the short chain fatty acid valproic acid (VPA). VPA is a drug used in humans primarily for epilepsy and seizure control, although it has been used in many nonepileptic conditions as well [10]. Based on several case studies and small-scale population-based studies in humans [11–16], in addition to mounting experimental evidence in animals [17–32], VPA has known teratogenicity and has long been suspected as a risk factor for ASD. This year, however, both a prospective study and a large-scale population-based study were published providing the most substantial evidence to date linking prenatal VPA exposure to an increased risk of ASD [33, 34]. Thus, in this paper we will review epidemiological evidence linking VPA to ASD and will discuss two promising leads in the study of ASD in light of human clinical findings and the valproic acid animal model of ASD. 2. Valproic Acid and ASD: Epidemiological Evidence Previous links between the antiepileptic drug VPA and ASD have been found and explored

References

[1]  T. Chomiak and B. Hu, “Alterations of neocortical development and maturation in autism: insight from valproic acid exposure and animal models of autism,” Neurotoxicology and Teratology, vol. 36, pp. 57–66, 2013.
[2]  E. Courchesne, K. Pierce, C. M. Schumann et al., “Mapping early brain development in autism,” Neuron, vol. 56, no. 2, pp. 399–413, 2007.
[3]  C. M. Schumann, C. S. Bloss, C. C. Barnes et al., “Longitudinal magnetic resonance imaging study of cortical development through early childhood in autism,” Journal of Neuroscience, vol. 30, no. 12, pp. 4419–4427, 2010.
[4]  D. G. Amaral, C. M. Schumann, and C. W. Nordahl, “Neuroanatomy of autism,” Trends in Neurosciences, vol. 31, no. 3, pp. 137–145, 2008.
[5]  R. Adolphs, “The neurobiology of social cognition,” Current Opinion in Neurobiology, vol. 11, no. 2, pp. 231–239, 2001.
[6]  H. C. Hazlett, M. D. Poe, G. Gerig et al., “Early brain overgrowth in autism associated with an increase in cortical surface area before age 2 years,” Archives of General Psychiatry, vol. 68, no. 5, pp. 467–476, 2011.
[7]  A. Raznahan, G. L. Wallace, L. Antezana, et al., “Compared to what? Early brain overgrowth in autism and the perils of population norms,” Biol Psychiatry, vol. 74, no. 8, pp. 563–575, 2013.
[8]  D. R. Morhardt, W. Barrow, M. Jaworski, and P. J. Accardo, “Head circumference in young children with autism: the impact of different head circumference charts,” Journal of Child Neurology, 2013.
[9]  C. Ecker, J. Suckling, S. C. Deoni et al., “Brain anatomy and its relationship to behavior in adults with autism spectrum disorder: a multicenter magnetic resonance imaging study,” Archives of General Psychiatry, vol. 69, no. 2, pp. 195–209, 2012.
[10]  J. A. Balfour and H. M. Bryson, “Valproic acid,” CNS Drugs, vol. 2, no. 2, pp. 144–173, 1994.
[11]  A. L. Christianson, N. Chesler, and J. G. R. Kromberg, “Fetal valproate syndrome: clinical and neuro-developmental features in two sibling pairs,” Developmental Medicine and Child Neurology, vol. 36, no. 4, pp. 361–369, 1994.
[12]  G. Williams, J. King, M. Cunningham, M. Stephan, B. Kerr, and J. H. Hersh, “Fetal valproate syndrome and autism: additional evidence of an association,” Developmental Medicine and Child Neurology, vol. 43, no. 3, pp. 202–206, 2001.
[13]  P. G. Williams and J. H. Hersh, “A male with fetal valproate syndrome and autism,” Developmental Medicine and Child Neurology, vol. 39, no. 9, pp. 632–634, 1997.
[14]  S. J. Moore, P. Turnpenny, A. Quinn et al., “A clinical study of 57 children with fetal anticonvulsant syndromes,” Journal of Medical Genetics, vol. 37, no. 7, pp. 489–497, 2000.
[15]  A. D. Rasalam, H. Hailey, J. H. G. Williams et al., “Characteristics of fetal anticonvulsant syndrome associated autistic disorder,” Developmental Medicine and Child Neurology, vol. 47, no. 8, pp. 551–555, 2005.
[16]  L. Laegreid, M. Kyllerman, T. Hedner, B. Hagberg, and G. Viggedahl, “Benzodiazepine amplification of valproate teratogenic effects in children of mothers with absence epilepsy,” Neuropediatrics, vol. 24, no. 2, pp. 88–92, 1993.
[17]  H. C. Lin, P. W. Gean, C.-C. Wang, Y. H. Chan, and P. Chen, “The amygdala excitatory/inhibitory balance in a valproate-induced rat autism model,” PLoS ONE, vol. 8, no. 1, Article ID e55248, 2013.
[18]  V. Bambini-Junior, L. Rodrigues, G. A. Behr, J. C. F. Moreira, R. Riesgo, and C. Gottfried, “Animal model of autism induced by prenatal exposure to valproate: behavioral changes and liver parameters,” Brain Research, vol. 1408, pp. 8–16, 2011.
[19]  P. E. Binkerd, J. M. Rowland, H. Nau, and A. G. Hendrickx, “Evaluation of valproic acid (VPA) developmental toxicity and pharmacokinetics in Sprague-Dawley rats,” Fundamental and Applied Toxicology, vol. 11, no. 3, pp. 485–493, 1988.
[20]  D. Dufour-Rainfray, P. Vourc'h, A. Le Guisquet et al., “Behavior and serotonergic disorders in rats exposed prenatally to valproate: a model for autism,” Neuroscience Letters, vol. 470, no. 1, pp. 55–59, 2010.
[21]  K. C. Kim, P. Kim, H. S. Go et al., “The critical period of valproate exposure to induce autistic symptoms in Sprague-Dawley rats,” Toxicology Letters, vol. 201, no. 2, pp. 137–142, 2011.
[22]  T. Rinaldi, K. Kulangara, K. Antoniello, and H. Markram, “Elevated NMDA receptor levels and enhanced postsynaptic long-term potentiation induced by prenatal exposure to valproic acid,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 33, pp. 13501–13506, 2007.
[23]  K. Markram, T. Rinaldi, D. L. Mendola, C. Sandi, and H. Markram, “Abnormal fear conditioning and amygdala processing in an animal model of autism,” Neuropsychopharmacology, vol. 33, no. 4, pp. 901–912, 2008.
[24]  F. I. Roullet, L. Wollaston, D. deCatanzaro, and J. A. Foster, “Behavioral and molecular changes in the mouse in response to prenatal exposure to the anti-epileptic drug valproic acid,” Neuroscience, vol. 170, no. 2, pp. 514–522, 2010.
[25]  T. Schneider and R. Przew?ocki, “Behavioral alterations in rats prenatally to valproic acid: animal model of autism,” Neuropsychopharmacology, vol. 30, no. 1, pp. 80–89, 2005.
[26]  T. Schneider, J. Turczak, and R. Przew?ocki, “Environmental enrichment reverses behavioral alterations in rats prenatally exposed to valproic acid: issues for a therapeutic approach in autism,” Neuropsychopharmacology, vol. 31, no. 1, pp. 36–46, 2006.
[27]  T. Schneider, A. Roman, A. Basta-Kaim et al., “Gender-specific behavioral and immunological alterations in an animal model of autism induced by prenatal exposure to valproic acid,” Psychoneuroendocrinology, vol. 33, no. 6, pp. 728–740, 2008.
[28]  C. V. Vorhees, “Behavioral teratogenicity of valproic acid: selective effects on behavior after prenatal exposure to rats,” Psychopharmacology, vol. 92, no. 2, pp. 173–179, 1987.
[29]  C. V. Vorhees, “Teratogenicity and developmental toxicity of valproic acid in rats,” Teratology, vol. 35, no. 2, pp. 195–202, 1987.
[30]  G. C. Wagner, K. R. Reuhl, M. Cheh, P. McRae, and A. K. Halladay, “A new neurobehavioral model of autism in mice: pre- and postnatal exposure to sodium valproate,” Journal of Autism and Developmental Disorders, vol. 36, no. 6, pp. 779–793, 2006.
[31]  A. C. Felix-Ortiz and M. Febo, “Gestational valproate alters BOLD activation in response to complex social and primary sensory stimuli,” PLoS ONE, vol. 7, no. 5, Article ID e37313, 2012.
[32]  R. Mychasiuk, S. Richards, A. Nakahashi, B. Kolb, and R. Gibb, “Effects of rat prenatal exposure to valproic acid on behaviour and neuro-anatomy,” Developmental Neuroscience, vol. 34, pp. 268–276, 2012.
[33]  R. L. Bromley, G. E. Mawer, M. Briggs, et al., “The prevalence of neurodevelopmental disorders in children prenatally exposed to antiepileptic drugs,” Journal of Neurology, Neurosurgery & Psychiatry, vol. 84, no. 6, pp. 637–643, 2013.
[34]  J. Christensen, T. K. Gr?nborg, M. J. S?rensen, et al., “Prenatal valproate exposure and risk of autism spectrum disorders and childhood autism,” Journal of the American Medical Association, vol. 309, no. 16, pp. 1696–1703, 2013.
[35]  J. C. Dean, H. Hailey, S. J. Moore, D. J. Lloyd, P. D. Turnpenny, and J. Little, “Long term health and neurodevelopment in children exposed to antiepileptic drugs before birth,” Journal of Medical Genetics, vol. 39, no. 4, pp. 251–259, 2002.
[36]  P. M. Rodier, J. L. Ingram, B. Tisdale, S. Nelson, and J. Romano, “Embryological origin for autism: developmental anomalies of the cranial nerve motor nuclei,” Journal of Comparative Neurology, vol. 370, no. 2, pp. 247–261, 1996.
[37]  P. M. Rodier, J. L. Ingram, B. Tisdale, and V. J. Croog, “Linking etiologies in humans and animal models: studies of autism,” Reproductive Toxicology, vol. 11, no. 2-3, pp. 417–422, 1997.
[38]  F. I. Roullet, J. K. Lai, and J. A. Foster, “In Utero exposure to valproic acid and autism—a current review of clinical and animal studies,” Neurotoxicology and Teratology, vol. 36, pp. 47–56, 2013.
[39]  D. Dufour-Rainfray, P. Vourc'h, S. Tourlet, D. Guilloteau, S. Chalon, and C. R. Andres, “Fetal exposure to teratogens: evidence of genes involved in autism,” Neuroscience and Biobehavioral Reviews, vol. 35, no. 5, pp. 1254–1265, 2011.
[40]  M. E. Bringas, F. N. Carvajal-Floresa, T. A. López-Ramírez, M. Atzori, and G. Flores, “Rearrangement of the dendritic morphology in limbic regions and altered exploratory behavior in a rat model of autism spectrum disorder,” Neuroscience, vol. 241, pp. 170–187, 2013.
[41]  K. Markram and H. Markram, “The intense world theory-a unifying theory of the neurobiology of autism,” Frontiers in Human Neuroscience, vol. 4, p. 224, 2010.
[42]  H. Markram, T. Rinaldi, and K. Markram, “The intense world syndrome-an alternative hypothesis for autism,” Frontiers in Neuroscience, vol. 1, no. 1, pp. 77–96, 2007.
[43]  C. Belzung, S. Leman, P. Vourc'h, and C. Andres, “Rodent models for autism: a critical review,” Drug Discovery Today, vol. 2, no. 2, pp. 93–101, 2005.
[44]  C. C. Woo and M. Leon, “Environmental enrichment as an effective treatment for autism: a randomized controlled trial,” Behavioral Neuroscience, vol. 127, no. 4, pp. 487–497, 2013.
[45]  S. E. Lazic and L. Essioux, “Improving basic and translational science by accounting for litter-to-litter variation in animal models,” BMC Neuroscience, vol. 14, p. 37, 2013.
[46]  A. Oguchi-Katayama, A. Monma, Y. Sekino, T. Moriguchi, and K. Sato, “Comparative gene expression analysis of the amygdala in autistic rat models produced by pre- and post-natal exposures to valproic acid,” Journal of Toxicological Sciences, vol. 38, no. 3, pp. 391–402, 2013.
[47]  S. Reynolds, A. Millette, and D. P. Devine, “Sensory and motor characterization in the postnatal valproate rat model of autism,” Developmental Neuroscience, vol. 34, no. 2-3, pp. 258–267, 2012.
[48]  C. L. Yochum, P. Dowling, K. R. Reuhl, G. C. Wagner, and X. Ming, “VPA-induced apoptosis and behavioral deficits in neonatal mice,” Brain Research, vol. 1203, pp. 126–132, 2008.
[49]  T. Chomiak, V. Karnik, E. Block, and B. Hu, “Altering the trajectory of early postnatal cortical development can lead to structural and behavioural features of autism,” BMC Neuroscience, vol. 11, p. 102, 2010.
[50]  E. Jager-Roman, A. Deichl, and S. Jakob, “Fetal growth, major malformations, and minor anomalies in infants born to women receiving valproic acid,” Journal of Pediatrics, vol. 108, no. 6, pp. 997–1004, 1986.
[51]  H. S. Go, K. C. Kim, C. S. Choi, et al., “Prenatal exposure to valproic acid increases the neural progenitor cell pool and induces macrocephaly in rat brain via a mechanism involving the GSK-3beta/beta-catenin pathway,” Neuropharmacology, vol. 63, no. 6, pp. 1028–1041, 2012.
[52]  M. W. Akhtar, J. Raingo, E. D. Nelson et al., “Histone deacetylases 1 and 2 form a developmental switch that controls excitatory synapse maturation and function,” Journal of Neuroscience, vol. 29, no. 25, pp. 8288–8297, 2009.
[53]  V. Balasubramaniyan, E. Boddeke, R. Bakels et al., “Effects of histone deacetylation inhibition on neuronal differentiation of embryonic mouse neural stem cells,” Neuroscience, vol. 143, no. 4, pp. 939–951, 2006.
[54]  J. Hsieh and F. H. Gage, “Epigenetic control of neural stem cell fate,” Current Opinion in Genetics and Development, vol. 14, no. 5, pp. 461–469, 2004.
[55]  J. Hsieh and F. H. Gage, “Chromatin remodeling in neural development and plasticity,” Current Opinion in Cell Biology, vol. 17, no. 6, pp. 664–671, 2005.
[56]  L. Sui and M. Chen, “Prenatal exposure to valproic acid enhances synaptic plasticity in the medial prefrontal cortex and fear memories,” Brain Research Bulletin, vol. 87, no. 6, pp. 556–563, 2012.
[57]  T. Rinaldi, C. Perrodin, and H. Markram, “Hyper-connectivity and hyper-plasticity in the medial prefrontal cortex in the valproic Acid animal model of autism,” Frontiers in Neural Circuits, vol. 2, p. 4, 2008.
[58]  M. Fukuchi, T. Nii, N. Ishimaru et al., “Valproic acid induces up- or down-regulation of gene expression responsible for the neuronal excitation and inhibition in rat cortical neurons through its epigenetic actions,” Neuroscience Research, vol. 65, no. 1, pp. 35–43, 2009.
[59]  B. Monti, E. Polazzi, and A. Contestabile, “Biochemical, molecular and epigenetic mechanisms of valproic acid neuroprotection,” Current Molecular Pharmacology, vol. 2, no. 1, pp. 95–109, 2009.
[60]  S. Jessberger, K. Nakashima, G. D. Clemenson Jr. et al., “Epigenetic modulation of seizure-induced neurogenesis and cognitive decline,” Journal of Neuroscience, vol. 27, no. 22, pp. 5967–5975, 2007.
[61]  S. Yasuda, M.-H. Liang, Z. Marinova, A. Yahyavi, and D.-M. Chuang, “The mood stabilizers lithium and valproate selectively activate the promoter IV of brain-derived neurotrophic factor in neurons,” Molecular Psychiatry, vol. 14, no. 1, pp. 51–59, 2009.
[62]  J. L. MacDonald and A. J. Roskams, “Histone deacetylases 1 and 2 are expressed at distinct stages of neuro-glial development,” Developmental Dynamics, vol. 237, no. 8, pp. 2256–2267, 2008.
[63]  A. G. Foley, S. Gannon, N. Rombach-Mullan, et al., “Class I histone deacetylase inhibition ameliorates social cognition and cell adhesion molecule plasticity deficits in a rodent model of autism spectrum disorder,” Neuropharmacology, vol. 63, no. 4, pp. 750–760, 2012.
[64]  C. J. Phiel, F. Zhang, E. Y. Huang, M. G. Guenther, M. A. Lazar, and P. S. Klein, “Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen,” Journal of Biological Chemistry, vol. 276, no. 39, pp. 36734–36741, 2001.
[65]  O. H. Kr?mer, P. Zhu, H. P. Ostendorff et al., “The histone deacetylase inhibitor valproic acid selectively induces proteasomal degradation of HDAC2,” EMBO Journal, vol. 22, no. 13, pp. 3411–3420, 2003.
[66]  Y. Hao, T. Creson, L. Zhang et al., “Mood stabilizer valproate promotes ERK pathway-dependent cortical neuronal growth and neurogenesis,” Journal of Neuroscience, vol. 24, no. 29, pp. 6590–6599, 2004.
[67]  C. F. Geier, K. Garver, R. Terwilliger, and B. Luna, “Development of working memory maintenance,” Journal of Neurophysiology, vol. 101, no. 1, pp. 84–99, 2009.
[68]  G. Golarai, D. G. Ghahremani, S. Whitfield-Gabrieli et al., “Differential development of high-level visual cortex correlates with category-specific recognition memory,” Nature Neuroscience, vol. 10, no. 4, pp. 512–522, 2007.
[69]  B. Luna, K. E. Garver, T. A. Urban, N. A. Lazar, and J. A. Sweeney, “Maturation of cognitive processes from late childhood to adulthood,” Child Development, vol. 75, no. 5, pp. 1357–1372, 2004.
[70]  K. S. Scherf, M. Behrmann, K. Humphreys, and B. Luna, “Visual category-selectivity for faces, places and objects emerges along different developmental trajectories,” Developmental Science, vol. 10, no. 4, pp. F15–F30, 2007.
[71]  P. Shaw, K. Eckstrand, W. Sharp et al., “Attention-deficit/hyperactivity disorder is characterized by a delay in cortical maturation,” Proceedings of the National Academy of Sciences of the United States of America, vol. 104, no. 49, pp. 19649–19654, 2007.
[72]  N. Gogtay, J. N. Giedd, L. Lusk et al., “Dynamic mapping of human cortical development during childhood through early adulthood,” Proceedings of the National Academy of Sciences of the United States of America, vol. 101, no. 21, pp. 8174–8179, 2004.
[73]  E. I. Knudsen, J. J. Heckman, J. L. Cameron, and J. P. Shonkoff, “Economic, neurobiological, and behavioral perspectives on building America's future workforce,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 27, pp. 10155–10162, 2006.
[74]  J. L. Elman, “Learning and development in neural networks: the importance of starting small,” Cognition, vol. 48, no. 1, pp. 71–99, 1993.
[75]  S. R. Quartz, “The constructivist brain,” Trends in Cognitive Sciences, vol. 3, no. 2, pp. 48–57, 1999.
[76]  P. Penzes, M. E. Cahill, K. A. Jones, J. Vanleeuwen, and K. M. Woolfrey, “Dendritic spine pathology in neuropsychiatric disorders,” Nature Neuroscience, vol. 14, no. 3, pp. 285–293, 2011.
[77]  N. Mosammaparast and Y. Shi, “Reversal of histone methylation: biochemical and molecular mechanisms of histone demethylases,” Annual Review of Biochemistry, vol. 79, pp. 155–179, 2010.
[78]  E. W. Tung and L. M. Winn, “Epigenetic modifications in valproic acid-induced teratogenesis,” Toxicology and Applied Pharmacology, vol. 248, no. 3, pp. 201–209, 2010.
[79]  A. E. Handel, G. C. Ebers, and S. V. Ramagopalan, “Epigenetics: molecular mechanisms and implications for disease,” Trends in Molecular Medicine, vol. 16, no. 1, pp. 7–16, 2010.
[80]  R. Tuchman and I. Rapin, “Epilepsy in autism,” Lancet Neurology, vol. 1, no. 6, pp. 352–358, 2002.
[81]  M. G. Chez, M. Chang, V. Krasne, C. Coughlan, M. Kominsky, and A. Schwartz, “Frequency of epileptiform EEG abnormalities in a sequential screening of autistic patients with no known clinical epilepsy from 1996 to 2005,” Epilepsy and Behavior, vol. 8, no. 1, pp. 267–271, 2006.
[82]  K. M. Dalton, B. M. Nacewicz, T. Johnstone et al., “Gaze fixation and the neural circuitry of face processing in autism,” Nature Neuroscience, vol. 8, no. 4, pp. 519–526, 2005.
[83]  C. W. Nordahl, R. Scholz, X. Yang et al., “Increased rate of amygdala growth in children aged 2 to 4 years with autism spectrum disorders: a longitudinal study,” Archives of General Psychiatry, vol. 69, no. 1, pp. 53–61, 2012.
[84]  K. C. Kim, D. K. Lee, H. S. Go, et al., “Pax6-dependent cortical glutamatergic neuronal differentiation regulates autism-like behavior in prenatally valproic acid-exposed rat offspring,” Molecular Neurobiology, 2013.
[85]  A. Banerjee, F. García-Oscos, S. Roychowdhury, et al., “Impairment of cortical GABAergic synaptic transmission in an environmental rat model of autism,” International Journal of Neuropsychopharmacology, vol. 16, no. 6, pp. 1309–1318, 2013.
[86]  L. Q. Uddin, K. Supekar, C. J. Lynch, et al., “Salience network-based classification and prediction of symptom severity in children with autism,” JAMA Psychiatry, vol. 70, no. 8, pp. 869–879, 2013.
[87]  Q. Chen, S. He, X. Hu et al., “Differential roles of NR2A- and NR2B-containing NMDA receptors in activity-dependent brain-derived neurotrophic factor gene regulation and limbic epileptogenesis,” Journal of Neuroscience, vol. 27, no. 3, pp. 542–552, 2007.
[88]  H. W. Horch and L. C. Katz, “BDNF release from single cells elicits local dendritic growth in nearby neurons,” Nature Neuroscience, vol. 5, no. 11, pp. 1177–1184, 2002.
[89]  A. K. McAllister, D. C. Lo, and L. C. Katz, “Neurotrophins regulate dendritic growth in developing visual cortex,” Neuron, vol. 15, no. 4, pp. 791–803, 1995.
[90]  A. K. McAllister, L. C. Katz, and D. C. Lo, “Neurotrophin regulation of cortical dendritic growth requires activity,” Neuron, vol. 17, no. 6, pp. 1057–1064, 1996.
[91]  D. K. Binder, S. D. Croll, C. M. Gall, and H. E. Scharfman, “BDNF and epilepsy: too much of a good thing?” Trends in Neurosciences, vol. 24, no. 1, pp. 47–53, 2001.
[92]  M. Stamou, K. M. Streifel, P. E. Goines, and P. J. Lein, “Neuronal connectivity as a convergent target of gene x environment interactions that confer risk for autism spectrum disorders,” Neurotoxicology and Teratology, vol. 36, pp. 3–16, 2013.
[93]  N. C. Spitzer, “Activity-dependent neurotransmitter respecification,” Nature Reviews Neuroscience, vol. 13, no. 2, pp. 94–106, 2012.
[94]  N. C. Spitzer, “Electrical activity in early neuronal development,” Nature, vol. 444, no. 7120, pp. 707–712, 2006.
[95]  J. L. Rubenstein and M. M. Merzenich, “Model of autism: increased ratio of excitation/inhibition in key neural systems,” Genes, Brain and Behavior, vol. 2, no. 5, pp. 255–267, 2003.
[96]  M. Vogelauer, A. S. Krall, M . A. McBrian, J.-Y. Li, and S. A. Kurdistani, “Stimulation of histone deacetylase activity by metabolites of intermediary metabolism,” Journal of Biological Chemistry, vol. 287, no. 38, pp. 32006–32016, 2012.
[97]  P. Rajendran, D. E. Williams, E. Ho, and R. H. Dashwood, “Metabolism as a key to histone deacetylase inhibition,” Critical Reviews in Biochemistry and Molecular Biology, vol. 46, no. 3, pp. 181–199, 2011.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413