Chapter Six - Epigenetic Mechanisms in Autism Spectrum Disorder

https://doi.org/10.1016/B978-0-12-801311-3.00006-8Get rights and content

Abstract

Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by impaired social interactions, language deficits, as well as restrictive or repetitive behaviors. ASD is clinically heterogeneous with a complex etiopathogenesis which may be conceptualized as a dynamic interplay between heterogeneous environmental cues and predisposing genetic factors involving complex epigenetic mechanisms. Inherited and de novo copy number variants provide novel information regarding genes contributing to ASD. Epigenetic marks are stable, yet potentially reversible, chromatin modifications that alter gene expression profiles by locally changing the degree of nucleosomal compaction, thereby opening or closing promoter access to the transcriptional machinery. Here, we review progress on studies designed to provide a better understanding of how epigenetic mechanisms impact transcriptional programs operative in the brain that contribute to ASD.

Introduction

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by symptoms that include deficits in social interactions, under-developed communication skills, and restrictive or repetitive behaviors. Population-wide prevalence of ASD is approximately 1% and it is more common in males than females (4:1) (Abrahams and Geschwind, 2008, Bailey et al., 1995, Baron-Cohen et al., 2009, O'Roak and State, 2008, Veenstra-Vanderweele and Cook, 2004). ASD is clinically and etiologically heterogeneous with many of the diagnostic symptoms showing considerable variation in severity. Approximately 5–20% of cases involve large effect de novo copy number variants. Genome-wide association studies are frequently used to compare the frequencies of single-nucleotide polymorphisms (SNPs) in ASD DNA samples. While no replicable single SNP variants have been identified, the cumulative contributions of inherited genetic variation over many small effect loci has recently been estimated to be as high as 40% (Klei et al., 2012).

Microarrays are used extensively to map the large number of copy number variants (CNVs) in the human population (Cook & Scherer, 2008). These mutations are variable in length and are either inherited or arise due to spontaneous (de novo) duplication, insertion, or deletion across the genome. Given the phenotypic overlap, it is not surprising that CNVs tend to be more frequent in patients with psychiatric disorders of neurodevelopmental origin such as intellectual disability/mental retardation (including patients with learning deficits), schizophrenia (SZ), and ASD (De Lacy and King, 2013, Guilmatre et al., 2009). Various tests of cognitive function show that control subjects known to be carrying CNVs that confer risk for either ASD or SZ test in terms of cognitive performance at a level intermediate between SZ and population controls (Stefansson et al., 2014). Moreover, CNVs do not all affect the same cognitive domains and vary considerably from one mutation to another (Stefansson et al., 2014). Two important goals of clinical geneticists are (1) to improve CNV detection and (2) to improve the phenotyping of CNV carriers that also exhibit psychotic symptoms. Recent analyses of topological networks derived from ASD CNVs and mouse functional genomics are being used to unveil highly detailed ASD-associated interaction networks that allow testing of novel hypotheses regarding cellular signaling and biological function (Noh et al., 2013, Veenstra-Vanderweele and Blakely, 2012).

One of the more frequently observed CNVs associated with high penetrance for ASD is the maternal duplication of chromosome 15q11-13. A second well-known CNV is a 600 kb microdeletion/microinsertion at chromosome 16p11.2 which occurs in some 1% of sporadic ASD cases (Cook & Scherer, 2008). Syndromic ASD, which occurs in 10% of cases, is observed when diagnostic behaviors are comorbid with a recognized syndrome. Some of the monogenic (syndromic) conditions associated with the ASD phenotype are Rett (RTT) syndrome (MECP2 gene; Amir et al., 1999), fragile X syndrome (FXS, FMR1), tuberous sclerosis (TSC1 and TSC2; Wiznitzer, 2004), neurofibromatosis (NF1), Timothy syndrome (CACNA1; Splawski et al., 2006), and cortical dysplasia–focal epilepsy syndrome (CNTNAP2; Strauss et al., 2006). Each of the above conditions exhibits phenotypes that overlap with ASD and hence offer important insight into how the corresponding genes contribute to pathogenesis.

Several twin studies have reported concordance rates between MZ twins at between 70% and 90%, while DZ twin concordance rates vary from 6% to 10%, with a more than 20-fold increased risk for siblings (Bailey et al., 1995, Eapen et al., 2013, Steffenburg et al., 1989). Phenotypic discordance between MZ twins is often associated with de novo mutations (Hallmayer et al., 2011, Zafeiriou et al., 2013) and numerous nonshared environmental factors including in utero growth restrictions (these include uneven blood supply, placental dysfunction, differential allelic expression, etc., and stochastic noise (Czyz, Morahan, Ebers, & Ramagopalan, 2012)). In addition, numerous pre and perinatal factors, e.g., low birth weight and prematurity, are among multiple factors associated with higher risk of ASD (Gardener, Spiegelman, & Buka, 2011). These observations are consistent with the hypothesis that environmental factors contribute to ASD and support an epigenetic component in phenotypic variation. This variation may be the consequence of parental allelic origin (imprinting) or the effects of DNA methylation/hydroxymethylation on the epigenome. Moreover, epigenetic factors also manifest at multiple additional levels including histone tail modifications, variations in methyl DNA-binding proteins, etc. Recently, there has been a growth in interest in studies of the involvement of epigenetic mechanisms in the pathophysiology of ASD. The overlapping phenotypic features of ASD that are shared with related neurodevelopmental disorders may be explained through altered gene expression profiles filtered through various epigenetic mechanisms (Lasalle, 2013, Miyauchi and Voineagu, 2013, Voineagu et al., 2011). In other words, as researchers begin to compare the gene expression networks between different neurodevelopmental disorders, differences in specific biological pathways and brain regions that reflect the phenotypes associated with each disease should become apparent (Hoerder-Suabedissen et al., 2013, Parikshak et al., 2013, Willsey et al., 2013). In summary, one plausible mechanistic approach to understanding the etiology of ASD supports the concept that environmental/epigenetic perturbations incurred during early nervous system development operate on and enhance the contributions of a large number of susceptibility genes identified as either inherited or de novo mutations which themselves may influence epigenetic regulation (see Fig. 6.1). Here, we summarize progress on epigenetic mechanisms operative in brain with the goal of understanding the distinct or overlapping features of these mechanisms in the etiopathogenesis of ASD. We provide a basic overview of histone modifications, DNA methylation, and hydroxymethylation before proceeding onto subsequent topics. While we have not discussed the impact of microRNAs (miRNAs), long noncoding RNAs (ncRNAs), and enhancer RNAs on transcriptional regulation, there have been several recent reviews on these subjects (Melios and Sur, 2012, Peschansky and Wahlestedt, 2014, Roberts et al., 2014, Velmeshev et al., 2013, Wilkinson and Campbell, 2013).

Section snippets

Histones

Histones are small basic proteins composed of a globular core, C-terminal and N-terminal tails. DNA is a double-stranded helix wrapped around a histone octamer composed of two copies each of histones H2a, H2b, H3, and H4 and 146 bp of superhelical DNA. This structure, called the nucleosome, is the fundamental unit of chromatin. Arrays of nucleosomes are organized along the chromosome in compact structures which facilitate extensive interactions between the chromatin and various nuclear

Genetic Defects with Epigenetic Implications

As noted in Section 1, phenotypic overlap with autism has been observed in several monogenic neurodevelopmental disorders, including FXS, RTT syndrome, MECP2 duplication syndrome (MDS), Down syndrome, Angelman syndrome (AS), Prader–Willi syndrome, Williams syndrome, fetal anticonvulsant syndrome, etc. Despite differences in the etiology of these conditions, the overlapping features evident on clinical presentation strongly suggest the possibility that common neurobiological pathways may be

GABAergic genes

A role for the GABAergic inhibitory system in the pathophysiology of ASD has been consistently reported based on the results of several postmortem human brain studies. These studies include reports of (1) reduction in the number of GABAergic Purkinje cells in cerebellar cortex (Bailey et al., 1998, Bauman and Kemper, 1985, Whitney et al., 2004), (2) reduction by ~ 50% of the GABA-synthesizing enzymes glutamic acid decarboxylase 65 (GAD2) in cerebellum and GAD1 in parietal cortex of ASD patients (

Environmental Model of Autism

With the increased prevalence of pervasive developmental disorders and ASD during the last several years, there have been ongoing efforts to obtain a better understanding of whether this represents a broadening of the diagnostic criteria or the underlying epidemiology. In addition to the impact of genetic risk, environmental/epigenetic factors interact with the genome during early development, playing an important role in determining risk upon clinical presentation (Chaste & Leboyer, 2012).

Conclusions

The epigenetic landscape represents a platform by which multiple environmental factors interact with complex genetic milieu, resulting in modifications in gene expression that shape cellular and neuronal functions, ultimately directing the specification of individual diversity. These epigenetic mechanisms are also, in part, responsible for shaping the ASD phenotype. Based on recent correlated network analysis of gene expression data, the focus of ASD research has been on genes whose products

Acknowledgments

This work was supported by a Lever Award from the Chicago Biomedical Consortium and NIH grants 1P50 MH094267 (E. H. C.), 1P50 HD055751 (E. H. C.), and 5 R01 MH093348-03 (A. G.). The authors would like to thank Myhidin Shehu, M.D., for help in preparing the figures.

References (245)

  • E.H. Cook et al.

    Linkage-disequilibrium mapping of autistic disorder, with 15q11-13 markers

    American Journal of Human Genetics

    (1998)
  • E. Costa et al.

    Dendritic spine hypoplasticity and downregulation of reelin and GABAergic tone in schizophrenia vulnerability

    Neurobiology of Disease

    (2001)
  • D. Dufour-Rainfray et al.

    Fetal exposure to teratogens: Evidence of genes involved in autism

    Neuroscience and Biobehavioral Reviews

    (2011)
  • M.G. Erlander et al.

    Two genes encode distinct glutamate decarboxylases

    Neuron

    (1991)
  • S.H. Fatemi et al.

    Glutamic acid decarboxylase 65 and 67 kDa proteins are reduced in autistic parietal and cerebellar cortices

    Biological Psychiatry

    (2002)
  • S.L. Fyffe et al.

    Deletion of Mecp2 in Sim1-expressing neurons reveals a critical role for MeCP2 in feeding behavior, aggression, and the response to stress

    Neuron

    (2008)
  • M. Gardiner-Garden et al.

    CpG islands in vertebrate genomes

    Journal of Molecular Biology

    (1987)
  • P.A. Ginno et al.

    R-loop formation is a distinctive characteristic of unmethylated human CpG island promoters

    Molecular Cell

    (2012)
  • A.J. Guastella et al.

    Intranasal oxytocin improves emotion recognition for youth with autism spectrum disorders

    Biological Psychiatry

    (2010)
  • A. Guidotti et al.

    Characterization of the action of antipsychotic subtypes on valproate-induced chromatin remodeling

    Trends in Pharmacological Sciences

    (2009)
  • M. Heinrichs et al.

    Oxytocin, vasopressin, and human social behavior

    Frontiers in Neuroendocrinology

    (2009)
  • M.I. Holst et al.

    Engrailed-2 regulates genes related to vesicle formation and transport in cerebellar Purkinje cells

    Molecular and Cellular Neurosciences

    (2008)
  • M.W. Abdallah et al.

    Neonatal levels of neurotrophic factors and risk of autism spectrum disorders

    Acta Psychiatrica Scandinavica

    (2013)
  • B.S. Abrahams et al.

    Advances in autism genetics: On the threshold of a new neurobiology

    Nature Reviews. Genetics

    (2008)
  • M. Adachi et al.

    MeCP2-mediated transcription repression in the basolateral amygdala may underlie heightened anxiety in a mouse model of Rett syndrome

    Journal of Neuroscience

    (2009)
  • T. Aid et al.

    Mouse and rat BDNF gene structure and expression revisited

    Journal of Neuroscience Research

    (2007)
  • U. Albrecht et al.

    Imprinted expression of the murine Angelman syndrome gene, Ube3a, in hippocampal and Purkinje neurons

    Nature Genetics

    (1997)
  • R.E. Amir et al.

    Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2

    Nature Genetics

    (1999)
  • E. Andari et al.

    Promoting social behavior with oxytocin in high-functioning autism spectrum disorders

    Proceedings of the National Academy of Sciences of the United States of America

    (2010)
  • A.E. Autry et al.

    Brain-derived neurotrophic factor and neuropsychiatric disorders

    Pharmacological Reviews

    (2012)
  • S.L. Baader et al.

    Ectopic overexpression of engrailed-2 in cerebellar Purkinje cells causes restricted cell loss and retarded external germinal layer development at lobule junctions

    Journal of Neuroscience

    (1998)
  • A. Bailey et al.

    Autism as a strongly genetic disorder: Evidence from a British twin study

    Psychological Medicine

    (1995)
  • A. Bailey et al.

    A clinicopathological study of autism

    Brain

    (1998)
  • J.M. Bandim et al.

    Autism and Möbius sequence: An exploratory study of children in northeastern Brazil

    Arquivos de Neuro-Psiquiatria

    (2003)
  • A.J. Bannister et al.

    Regulation of chromatin by histone modifications

    Cell Research

    (2011)
  • S. Baron-Cohen et al.

    Prevalence of autism-spectrum conditions: UK school-based population study

    British Journal of Psychiatry

    (2009)
  • G. Barreto et al.

    Gadd45a promotes epigenetic gene activation by repair-mediated DNA demethylation

    Nature

    (2007)
  • T. Batsukh et al.

    CHD8 interacts with CHD7, a protein which is mutated in CHARGE syndrome

    Human Molecular Genetics

    (2010)
  • M. Bauman et al.

    Histoanatomic observations of the brain in early infantile autism

    Neurology

    (1985)
  • E. Ben-David et al.

    Combined analysis of exome sequencing points toward a major role for transcription regulation during brain development in autism

    Molecular Psychiatry

    (2013)
  • A.T. Berg et al.

    Risk and correlates of autism spectrum disorder in children with epilepsy: A community-based study

    Journal of Child Neurology

    (2011)
  • S. Beri et al.

    DNA methylation regulates tissue-specific expression of Shank3

    Journal of Neurochemistry

    (2007)
  • R.J. Berry

    Maternal prenatal folic acid supplementation is associated with a reduction in development of autistic disorder

    Journal of Pediatrics

    (2013)
  • T.H. Bestor

    DNA methylation: Evolution of a bacterial immune function into a regulator of gene expression and genome structure in higher eukaryotes

    Philosophical Transactions of the Royal Society of London B

    (1990)
  • P. Bittigau et al.

    Antiepileptic drugs and apoptotic neurodegeneration in the developing brain

    Proceedings of the National Academy of Sciences of the United States of America

    (2002)
  • G.J. Blatt et al.

    Density and distribution of hippocampal neurotransmitter receptors in autism: An autoradiographic study

    Journal of Autism and Developmental Disorders

    (2001)
  • R.L. Bromley et al.

    The prevalence of neurodevelopmental disorders in children prenatally exposed to antiepileptic drugs

    Journal of Neurology, Neurosurgery, and Psychiatry

    (2013)
  • R.L. Bromley et al.

    Autism spectrum disorders following in utero exposure to antiepileptic drugs

    Neurology

    (2008)
  • G. Cavalli et al.

    Functional implications of genome topology

    Nature Structural & Molecular Biology

    (2013)
  • M. Chahrour et al.

    MeCP2, a key contributor to neurological disease, activates and represses transcription

    Science

    (2008)
  • Cited by (0)

    View full text