Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Aug 1.
Published in final edited form as: Mol Psychiatry. 2013 Sep 3;19(8):862–871. doi: 10.1038/mp.2013.114

Common DNA methylation alterations in multiple brain regions in autism

Christine Ladd-Acosta 1,2, Kasper D Hansen 2,3, Eirikur Briem 2,4, M Daniele Fallin 1,2, Walter E Kaufmann 5,*, Andrew P Feinberg 2,4
PMCID: PMC4184909  NIHMSID: NIHMS630368  PMID: 23999529

Abstract

Autism spectrum disorders (ASD) are increasingly common neurodevelopmental disorders defined clinically by a triad of features including impairment in social interaction, impairment in communication in social situations, and restricted and repetitive patterns of behavior and interests, with considerable phenotypic heterogeneity among individuals. Although heritability estimates for ASD are high, conventional genetic-based efforts to identify genes involved in ASD have yielded only few reproducible candidate genes that account for only a small proportion of ASDs. There is mounting evidence to suggest environmental and epigenetic factors play a stronger role in the etiology of ASD than previously thought. To begin to understand the contribution of epigenetics to ASD, we have examined DNA methylation (DNAm) in a pilot study of post-mortem brain tissue from 19 autism cases and 21 unrelated controls, among three brain regions including dorsolateral prefrontal cortex, temporal cortex, and cerebellum. We measured over 485,000 CpG loci across a diverse set of functionally relevant genomic regions using the Infinium HumanMethylation450 BeadChip and identified 4 genome-wide significant differentially methylated regions (DMRs) using a novel bumphunting approach and a permutation-based multiple testing correction method. We replicated 3/4 DMRs identified in our genome-wide screen in a different set of samples and across different brain regions. The DMRs identified in this study represent suggestive evidence for commonly altered methylation sites in ASD and provide several promising new candidate genes.

Keywords: DNA methylation, autism, epigenome, brain, 450k

INTRODUCTION

Autism Spectrum Disorders (ASD) are common neurodevelopmental disorders defined by three core clinical features: (1) impairment in social interaction; (2) impairment in communication in social situations; and (3) restricted and repetitive patterns of behaviors and interests1, 2. Given the striking recent increase in ASD prevalence and its associated social and economic impact3, there has been considerable interest in understanding the biological basis of ASD4; however the molecular underpinnings of ASD still remain elusive.

Familial aggregation, rare chromosomal abnormality, and twin studies suggest there is a strong genetic basis to ASD. Numerous genetic studies have examined common variants, rare mutations, and copy number variants (CNVs) across many thousands of autistic children but have had limited success. The strongest genetic candidates identified to date involve rare mutations. For example, rare variants have been identified in only 6/461 ASD cases for CACNA1H5, 6 and CNTN4 deletions have been identified in 7/~2000 cases710. These genes represent two of the five leading candidate genes in ASD to date11. Several promising recent studies have revealed an association between de novo mutations and CNVs with ASD; however de novo events do not directly explain the high heritability OF ASD12. A recent review suggests that at most 10–15% of all non-syndromic cases of ASD are associated with genetic alterations13, and to date, whole-genome and in-depth exon sequencing studies have suggested ASD is minimally explained by genetics alone.

Although epigenetic involvement has been a persistent minority view of ASD etiology, there is mounting evidence to suggest the contribution of epigenetics to ASD is stronger than previously thought. Direct evidence14 comes from identification of ASD-associated chromosomal abnormalities in imprinted regions, ASD linkage and association in areas of known imprinting, and evidence of parent-of-origin effects in association and linkage signals15, 16. Additional support stems from the observation of autistic features, sometimes reaching the diagnostic threshold, in individuals affected with known epigenetic disorders such as Angelman, fragile X, and Rett Syndromes. Rett Syndrome is caused by mutations in the MECP217, a gene that encodes a protein responsible for recognizing DNA methylation marks throughout the genome. Angelman syndrome involves alterations of the chromosome 15q11-q13 imprinted gene cluster18. Lastly, fragile X syndrome is caused by the expansion of a methylated CGG repeat in the 5′ untranslated region of the FMR1 gene19, resulting in inappropriate gene silencing.

Previous genome-wide DNA methylation (DNAm) assays, applied to ASD20, 21 and other diseases, have focused on measuring DNAm at CpG sites assumed to be functionally important, i.e. CpG islands (CGI) and promoters. However, we recently demonstrated that most functionally important changes in DNAm occur outside of CGI and promoter regions, specifically at CpG island shores22, 23. Non-island and promoter regions are the predominant sites of aberrant DNAm for many diseases including several types of cancers 22, 2428 and rheumatoid arthritis29, 30. Previous limited studies of specific candidate genes and CpG island or promoter arrays have shown epigenetic differences in lymphoblastoid cell lines and brain samples from individuals with ASD20, 21. However, these studies were limited either in scope, focused on particular regions of the genome, or by sample size, and/or by cell type (lymphoblast cell lines). One recent study that examined trimethylated histone 3 lysine 4 (H3K4me3) marks in post-mortem prefrontal cortex brain tissue identified a slight spreading of H3K4me3 marks, normally restricted to promoter regions, into gene bodies among 4/16 autistic individuals31. Nonetheless, to date, no study has examined DNAm on a genome-scale, not limited to CpG islands, in brain samples of ASD cases.

Here, we report the first genome-wide examination of DNAm in ASD among 40 post-mortem brain samples, including 19 cases and 21 controls, across 3 brain regions, using the Infinium HumanMethylation450 BeadChip (450K). Availability of ASD brain samples is extremely limited32, with the numbers examined in this study being comparable to other molecular analyses of ASD post-mortem brain samples31, 33. The 450K quantitatively measures DNAm at 485,577 genomic loci at high value content regions including all CpG island and promoter regions as well as CpG shores, cancer and tissue-differentially methylated regions (DMRs), non-coding RNAs, and DNase hypersensitive sites34, 35. We applied a new methodology termed “bump hunting”36 to our analysis which allows effective modeling of measurement error, detection of differentially methylated regions (DMRs), and assessment of genome-wide statistical significance (via permutation testing). Identifying regions of differential methylation is advantageous to single site analysis for multiple reasons including better protection from technical artifacts associated with individual probes and functional significance of regional methylation change versus at a single CpG.

MATERIALS AND METHODS

Human post-mortem brain tissue samples

We acquired 41 post-mortem brain tissue samples, including 20 cases and 21 controls, representing three brain regions: temporal cortex (TC, n=16), prefrontal cortex (PFC, n=12), and cerebellum (CBL, n=13) from the Autism Tissue Program and Harvard Brain Tissue Resource Center at McLean Hospital, Belmont, MA (TC and PFC samples) and the National Institute of Child Health and Human Development (NICHD) Brain and Tissue Bank for Developmental Disorders at the University of Maryland, Baltimore (CBL samples). These 41 samples represent 36 individuals; 5 individuals have both a TC and PFC sample included in our analyses (Figure 1a). For each brain region, autistic and control samples were matched as best as possible for age, sex, and post-mortem interval; no significant differences were detected (t-test, p < 0.05 and Supplementary Table 1). Furthermore, we examined the relationship between DNAm and age/PMI for all 4 DMRs identified in our genomic screen; no correlation between the covariates and DNAm was observed (Supplementary Figures 1 and 2). All autistic individuals included in our analysis were diagnosed using scores from the Autism Diagnostic Interview (ADI-R) and/or the Autism Diagnostic Observation Schedule (ADOS). In Supplementary Table 2, we provide a list of post-mortem brain samples and their associated features.

Figure 1.

Figure 1

(a) Summary of post-mortem brain samples and analytic procedures utilized. (b and c) Modified region-based (“Dry”) Manhattan plots for epigenome-based analysis. Region-specific DNA methylation was calculated from 450K data, and p values were determined by permutation analysis to accommodate the genome-wide screen. (b) Three differentially methylated regions (DMRs) in the temporal cortex brain region are significantly associated with ASD. (c) One DMR in the cerebellum is significantly associated with ASD. Each red segment represents the adjusted p-value for a genomic region on the Illumina Infinium 450K Methylation BeadChip. The significance threshold was based on a family-wise error rate of 0.1.

Illumina Infinium 450K methylation measurements

Genomic DNA was extracted from TC and PFC samples using Trizol (Invitrogen) and from CBL samples using the MasterPure DNA purification kit (Epicentre Biotechnologies) according to the manufacturer’s specifications for each kit. Each DNA sample was bisulfite treated, 500ng of gDNA, using the EZ DNA methylation kit (Zymo Research) according to the manufacturers specifications, optimized for the 450K. All of the samples per brain region were processed on the same plate with cases and controls randomized across the wells to minimize potential plate or batch effects.

Statistical analysis

Overview and quality control measures

All statistical analyses were performed using R 2.15 and Bioconductor 2.9. Raw intensity files (.idat) were obtained and processed using minfi package37 to obtain log ratios of methylation percentage (M-values). We applied several quality control measures to remove any spurious samples or probes. First, we examined 450K control probes to assess bisulfite conversion, extension, hybridization, staining, specificity, negative control, and others; no outlier samples were detected. Next, we checked for sex-discrepancies by comparing self-reported genders against data derived sex values. No sex-discrepancies were identified. We then looked for poorly performing arrays by calculating the total array intensity (sum of the methylated and unmethylated signals across all probes on the array) for each sample; no substantial differences were detected. Unsupervised clustering of samples identified one sample that was inappropriately clustering with a different brain region (PFC sample clustering with CBL samples); this sample was removed from downstream analysis. We removed probes that had an annotated SNP (dbSNP134) at the single base extension or CpG site since it is possible that SNP differences in these locations may manifest as differential methylation on the 450K, leaving a total of 428,526 probes.

Preprocessing and normalization

The signal in the methylated and unmethylated channels was first computed without background correction. Each channel was normalized separately, by first quantile normalizing the signal for all autosomal probes and second by quantile normalizing the signal for the sex chromosomes for each sex separately. After normalization, log ratios of methylation percentages were computed (M-values). Previous work38 has shown M-values to perform better than Beta-values for Illumina methylation arrays.

Identification of differentially methylated regions (DMRs)

We adapted the bump hunting technique previously described36 to the 450K. Probes were assigned to clusters so that two neighboring probes in the same cluster are separated by at most 500bp. For each probe, we estimated the difference in average log-ratios between cases and controls, controlled for sex, and smoothed these estimated differences using running medians. Next, the smoothed estimated differences were thresholded based on the 97.5% percent quantile of the empirical distribution of the smoothed estimated differences. This yielded a set of putative DMRs. Significance was assigned by permutation testing. For each of 1000 permutations of case-control status, a new list of putative DMRs was obtained. The genome-wide family-wise error rate for each observed DMR was calculated as the proportion of null-derived DMRs across the genome with more CpGs and greater difference between cases and controls than the observed DMR. Note that most of the genome is not inside a DMR, and is therefore not assigned an empirical significance via this approach.

Replication analysis

For the 3 DMRs identified in our genome-wide screen of TC tissue, we performed replication analyses in PFC and CBL tissues using methylation data obtained using the 450K. For replication purposes, the PFC and CBL data was restricted to include only the 74 probes that were located within the 3 significant TC DMRs. Similarly, for the DMR we identified in CBL tissue, we performed replication analyses by restricting our analysis in PFC and TC tissues to the 12 sites located within the cerebellum DMR. P values were computed using a two-sided t-test with a significance threshold of 0.05. Any DMR identified in our genome-wide screen that also had at least 2 significant differentially methylated probes in a replication set was deemed a positive replication.

Assessment of cell composition differences

Brain cell type specific DNAm data and cell epigenotype specific (CETS) patterns 39 were acquired using the CETS package (http://psychiatry.igm.jhmi.edu/kaminsky/software.htm) in R/Bioconductor. Overlap between the 450K probes identified in our brain samples and the top 10,000 CETS marks for brain cell types was computed by simply comparing the two lists of 450K probe names. Using the CETS package, we then estimated the proportion of neuronal and glial cells for each post-mortem brain tissue sample in our study, as specified previously39.

RESULTS

Figure 1a depicts an overview of our experimental design and the brain samples utilized in our analyses. Briefly, we performed individual methylome analyses for each of three brain regions: dorsolateral prefrontal cortex (PFC, 6 cases/5 controls), temporal cortex (TC, 6 cases/10 controls), and cerebellum (CBL, 7 cases/6 controls) using a “bump hunting” approach.

Genome-wide screen

For each of the three brain regions we performed a genome-wide screen and identified a total of four genome-wide significant (adjusted p < 0.1) differentially methylated regions (DMRs), three in the superior temporal cortex and one in the cerebellum (Figure 1b, c and Table 1). Because bump-hunting identifies a differentially methylated region (DMR) rather than a differentially methylated position (DMP), the data display is similar but with a somewhat more austere appearance than a conventional Manhattan plot. Therefore, we have termed it a “dry Manhattan” plot suitable for this type of DNAm analysis. As described in detail in the Methods section, empirical P values reported in our dry Manhattan plots (Figure 1b, c) were calculated by permutation with a family-wise error rate of 0.1.

Table 1.

Genome-wide significant DMRs associated with ASD

Brain region Genomic location Nearest gene Gene distance (bp) ΔMa Adjusted p-valueb Replication (p < 0.05)c
TC PFC CB
TC chr6: 32115964-32117379 PRRT1 0 −7.8% 0.001 na yes yes
TC chr11: 2321770-2323272 C11orf21 0 −6.6% 0.013 na yes no
TC chr6: 29648379-29649092 ZFP57 3449 13.9% 0.019 na no yes*
CB chr5: 1594021-1595048 SDHAP3 0 15.8% 0.073 no no na

Abbreviations: TC, temporal cortex; PFC, prefrontal cortex; CB, cerebellum; M, DNA methylation

a

Mean methylation difference between autism and control groups for the differentially methylated region. Positive values reflect relative hypermethylation and negative values relative hypomethylation in autistic individuals compared to controls.

b

Empirical significance value, computed using permutation testing (n=1000).

c

Replication is equal to “yes” if any Illumina Infinium 450K probes located within the DMR (identified in the genome-screen) have a nominal p-value less than 0.05 in a different brain tissue, otherwise it is reported as “no”.

*

Sex-specific replication. Significance is not reached when males and females are examined as one set.

As shown in Figure 2a, we identified a DMR at the 3′ UTR of PRRT1, proline-rich transmembrane protein 1, with relative hypomethylation in the temporal cortex tissue of autistic individuals (shown in green) compared to controls (shown in purple). On average, the cases are 7.8% less methylated at this DMR (empirical P = 0.001) than controls (Table 1). It is possible that changes in copy number variants (CNVs) may manifest as DNAm changes on the 450K. We would expect CNVs to result in different total intensities, i.e. the sum of the methylated and unmethylated channels, on the 450K platform. Therefore to assess potential CNV changes, in the second panel of Figure 2a, we plot the difference between the total probe intensity for a given individual and the mean total intensity across all individuals for a given probe, with purple and green points denoting control and ASD, respectively. There are no clear differences between the case and control groups with respect to CNV status; thus, it is likely the DMR does not reflect a CNV.

Figure 2.

Figure 2

Two genome-wide significant differentially methylated regions (DMRs) located at PRRT1 (ac) and C11orf21/TSPAN32 (df) are relatively hypomethylated in temporal cortex tissue from autistic individuals. (a and d) DMR plots for temporal cortex brain samples. (b and e) DMR plots for prefrontal cortex brain samples. (c and f) DMR plots for cerebellum brain samples. For (a and d), methylation data is shown in the top panels. Smoothed lines denote mean methylation levels for individuals with (green) and without (purple) ASD across the DMR. Each point represents the methylation level of a particular individual, green for those with autism and purple for those without autism, at a specific genomic location. The methylation values range from 0 to 1 with 0 equal to 0% methylation and 1 equal to 100% methylation. The upper middle panels provide information about potential copy number changes. Each point, green for autistic and purple for normal individuals, represents the difference between total probe intensity for a given individual and the mean total intensity across all individuals for a given probe. The black points in the lower middle panel are individual probe-based nominal p-values for differences in methylation between the two groups (autism and normal). The dashed magenta line indicates the significance threshold used (nominal p < 0.05) such that points above the line are significant in replication analyses. The bottom panel displays relevant genome annotation information. For (b, c, e, and f), plots are as described above but blue and orange points represent individuals with and without autism, respectively. Dotted boxes describe the location of methylation values that correspond to probes with significant nominal p-values.

We also identified a DMR (empirical P=0.013) that is on average 6.6% less methylated in ASD temporal cortex tissue, shown in green, compared to controls, shown in purple (Table 1 and Figure 2b). This DMR is located within the promoter regions of tetraspanin 32 (TSPAN32), and chromosome 11 open reading frame 21 (C11orf21), and continues into the gene body of C11orf21 (bottom panel of Figure 2b). We do not observe substantial changes in CNV levels between cases and controls for this DMR (second panel of Figure 2b).

The third DMR (empirical P=0.019) in temporal cortex tissue is on average 13.9% more methylated in ASD, shown in green, compared to controls, shown in purple (Table 1 and Figure 3). The relatively hypermethylated DMR is located in an intergenic region; the nearest gene is ZFP57, located 3.5kb away (Figure 3a). Although a few control individuals appear to have a slight increase in copy number for a few probes, across the region there is no substantial difference in CNV state between cases and controls (Figure 3a, second panel).

Figure 3.

Figure 3

A genome-wide significant differentially methylated region (DMR) located 3.45 Kb upstream of ZNF57 is relatively hypermethylated in temporal cortex tissue from autistic individuals. (a) DMR plots for temporal cortex brain samples. (b) DMR plots for temporal cortex brain samples from male individuals. (c) DMR plots for temporal cortex brain samples from female individuals. For (a–c), methylation data is shown in the top panel. Lines denote mean methylation levels for individuals with (green) and without (purple) autism across the DMR. Each point represents the methylation level of a particular individual, green for those with autism and purple for those without autism, at a specific genomic location. The methylation values range from 0 to 1 with 0 equal to 0% methylation and 1 equal to 100% methylation. The upper middle panel provides information about potential copy number changes. Each point, green for autistic and purple for normal individuals, represents the difference between total probe intensity for a given individual and the mean total intensity across all individuals for a given probe. The black points in the lower middle panel are individual probe-based nominal p-values for differences in methylation between the two groups (autism and normal). The dashed magenta line indicates the significance threshold used (nominal p < 0.05) such that points above the line are significant in replication analyses. The bottom panel displays relevant genome annotation information. (d) DMR plots for all prefrontal cortex brain samples. (e) DMR plots for prefrontal cortex tissue samples from male individuals. (f) DMR plots for prefrontal cortex tissue samples from female individuals. (g) DMR plots for all cerebellum brain samples. (h) DMR plots for cerebellum samples from male individuals. (i) DMR plots for cerebellum samples from female individuals. For (d–h), plots are as described above but blue and orange points represent individuals with and without autism, respectively. Dotted boxes describe the location of methylation values that correspond to probes with significant nominal p-values.

Among 13 cerebellum samples, including 7 cases and 6 controls, we identified 1 significant DMR located within SDHAP3, succinate dehydrogenase complex, subunit A, flavoprotein pseudogene 3. Cases are, on average, 15.8% more methylated than controls across this region (Table 1), although there is considerable variation among the ASD group that seems to be, at least in part, related to CNV (Figure 4a). The cases with higher methylation levels also have higher total intensities, representing potential CNVs. Thus, the methylation change detected in this region may be influenced by an underlying CNV change. Nonetheless, it still implicates this particular region as important in the etiology of ASD.

Figure 4.

Figure 4

A genome-wide significant differentially methylated region (DMR) at SDHAP3 is relatively hypermethylated in cerebellum tissue from autistic individuals. (a) DMR plots for cerebellum brain samples. (b) DMR plots for prefrontal cortex brain samples. (c) DMR plots for temporal cortex brain samples. For (a), methylation data is shown in the top panel. Lines denote mean methylation levels for individuals with (green) and without (purple) autism across the DMR. Each point represents the methylation level of a particular individual, green for those with autism and purple for those without autism, at a specific genomic location. The methylation values range from 0 to 1 with 0 equal to 0% methylation and 1 equal to 100% methylation. The upper middle panel provides information about potential copy number changes. Each point, green for autistic and purple for normal individuals, represents the difference between total probe intensity for a given individual and the mean total intensity across all individuals for a given probe. The black points in the lower middle panel are individual probe-based nominal p-values for differences in methylation between the two groups (autism and normal). The dashed magenta line indicates the significance threshold used (nominal p < 0.05) such that points above the line are significant in replication analyses. The bottom panel displays relevant genome annotation information. For (b and c), plots are as described above but blue and orange points represent individuals with and without autism, respectively. Dotted boxes describe the location of methylation values that correspond to probes with significant nominal p-values.

Replication analyses

Finally, we sought to confirm DMRs identified in our genome-wide screen in an independent set of samples. In total 74 probes are located within the 3 TC DMRs and 12 probes are located within the 1 cerebellar DMR. Given the limited availability of post-mortem brain samples from autistic individuals, we considered replication of TC DMRs via evidence of association at those probes in the PFC and CBL tissue samples, and used the TC and PFC sample sets to replicate our CBL DMR finding. Since the samples for each brain region were obtained from different individuals, with the exception of 5 individuals that are represented in both the TC and PFC sample sets, this design is suitable to evaluate how well our genome-wide findings replicate in different individuals and across different brain regions. Figure 1 provides a summary of the total number of individuals and probes examined in our replication analyses.

For the PRRT1 associated DMR discovered in TC, we found significant methylation differences between cases and controls in PFC tissue for 14 of 33 sites (Figure 2b, blue). For clarity, we use dashed boxes to highlight methylation values that correspond to probes with significant p-values. We did not observe any differences related to copy number for this region in PFC samples (upper middle, Figure 2b). We also observed two significant CpG sites and others with suggestive significance among the cerebellar tissue (Figure 2c) in an independent sample of 7 cases and 6 controls. Consistent with both our genome-wide screen and PFC replication results, the cerebellum tissue from autistic individuals showed less methylation than control individuals at the PRRT1 locus (Figure 2c) and no differences in CNV between cases and controls.

Replication analysis of the TC DMR associated with TSPAN32 and C11orf21 revealed 7 of 26 and 1 of 26 significant (P < 0.05, third panel of Figures 2e and f) differentially methylated loci in the PFC and cerebellum tissue samples, respectively (Figures 2e and f). However, since we would expect to see one significant difference by chance, we do not report the CBL as significant in our replication result (Table 1, Figure 1a). As shown in the top panels of Figures 2e and 2f, autistic individuals (blue points) are less methylated than the controls (orange points) for both the PFC and CBL tissues, consistent with our genome-wide result in TC tissue. In addition, no CNV changes are observed at the significant loci (second panel, Figures 2e, f).

The third DMR identified from TC, located 3.5kb upstream of ZFP57, showed sex-specific replication in the CBL samples (Figure 3i). We decided to stratify by sex for this DMR because we observed two clusters of methylation values that were correlated with sex. Interestingly, for both TC and CBL tissues, upon stratification by sex we found that normal females are less methylated than autistic females. Since there was only one female, we could not assess significant differences between cases and controls for females in PFC tissue. However, in the one female (autistic) with PFC data, we observed high methylation levels (Figure 3f); consistent with our findings in the other 2 regions for autistic females (Figures 3c, i). We also observed suggestive evidence that autistic males have more DNAm variability in TC and CBL tissues than control individuals (Figures 3b, h) for this genomic region.

Lastly, although none of the loci located in the CBL DMR in SDHAP3 reached statistical significance in the replication sets (Figures 4b, c), we observed the same trend in PFC as we did in CBL. More specifically, a few autistic samples in PFC show methylation levels near 50% that are potentially related to CNV changes (Figure 4b).

Brain tissue heterogeneity

We evaluated the possibility that our ASD-associated DMRs may reflect underlying cell type heterogeneity between ASD cases and controls using neuronal and glial cell type specific methylation data 39. None of the 86 probes located within the ASD DMRs we identify are among the list of 10,000 probes with neuron-specific methylation values from a recent report 39. Furthermore, for each of our brain samples, we computed the proportion of neuronal and glial cells using the Guintivano et al. 39 algorithm and found no significant differences between the ASD cases and controls for either brain region (Supplementary Table 3). Thus, our findings do not simply reflect a shift in the proportion of neuronal and glial cells between ASD case and control samples.

DISCUSSION

We provide the first evidence for significant DNAm changes in post-mortem brain tissue from ASD patients. After adjusting for multiple testing, we identified a total of four differentially methylated regions, three in TC tissues and one in CBL. Three of the four DMRs identified via genome-wide screen replicated in independent samples from different brain regions. Although the fourth DMR did not meet significance criteria for replication, it did show the same trend in the PFC samples as we observed in our genome-wide screen using CBL samples. Lastly, we demonstrated that the DMRs we identified are not related to underlying brain tissue cell composition differences between ASD and control individuals.

There is substantial evidence to support the biological plausibility of the regions we identified in the etiology of ASD. The DMR within the PRRT1 3′ UTR overlaps two DNase hypersensitive sites and an alternative transcript finish site. Given the location it is possible that this DMR is an important regulatory site40, 41. Although little is known about the function or expression patterns of PRRT1 in humans it has been shown to be specifically expressed in the hippocampus of marmoset42, a nonhuman primate often used as an ideal model for a wide-variety of central nervous system disorders. The hippocampus brain region has been previously implicated in human studies of ASD demonstrating macroscopic and microscopic anatomical differences4346 and altered synaptic function and plasticity47 in autistic individuals. Mutations in other genes in the PRRT family have been shown to cause several neurological disorders in humans, some of them developmental in nature, including familial infantile seizures48, paroxysmal kinesigenic dyskinesia (PKD)49, hemiplegic migraine50, PKD combined with infantile seizures (ICCA)51, 52, and benign familial infantile seizures (BFIS)53, 54.

The second DMR identified in TC tissue is located in the promoter regions of TSPAN32 and C11orf21 and extends into the gene body of C11orf21. While there is no literature to describe the function of C11of21, TSPAN32 is important in cellular immunity55 and acts as a structural and cell signaling scaffold protein56. Functional mutations in other tetraspanins have been identified in schizophrenia and bipolar pateints 57. Mutations in several other scaffolding proteins such as SHANK35860, SHANK261, and NBEA62, have been identified in autistic individuals. Interestingly, this DMR is also located within an imprinted region of the genome. Imprinting 63 and parent-of-origin specific effects15, 16 have been previously implicated in ASD.

The third DMR is located about 3.5kb upstream of ZFP57 and overlaps the 5′ end of an alternatively spliced EST. ZFP57 is instrumental in maintaining imprinting marks during development64, 65 by providing a mechanism for targeting DNA methyltransferase66, responsible for transferring methyl groups to cytosines, to specific locations in the genome67. This is directly relevant to ASD since other methylation machinery proteins are mutated in neurodevelopmental syndromes associated with ASD, e.g. Rett Syndrome17.

Lastly, in cerebellar tissue we identified a DMR at an alternative promoter for SDHAP3, which is associated with a non-coding RNA and a small coding RNA. This DMR falls directly on a CTCF binding site and an active regulatory element site identified by the ENCODE project68. Thus, it is likely to be an important regulatory site; however, currently there is no literature describing the function of this particular gene. SDHAP3 is a member of the succinate dehydrogenase gene family that includes genes that are critical components of the metabolic machinery. Mitochondrial respiratory chain complex protein dysfunction has been previously associated with ASD6971.

For one of the DMRs, near ZPF57, we identified a striking difference in methylation between autistic and control females in cerebellar tissue. We performed a BLAST search of the DNA sequence for this DMR and did not find alignment to any other regions of the genome, including sex-chromosomes. While intriguing, we examined a relatively small number of female samples in our analyses and the biological significance of sexually dimorphic DNAm differences in the brain is unclear at this time. This result suggests future epigenetic studies may benefit from inclusion of females and additional sex-specific analyses.

While this study is the first to identify commonly altered DNAm in brain tissue from autistic individuals, we recognize there are several limitations that need to be carefully considered. First, from an epidemiological and statistical perspective we examined a relatively small number of samples, 40 in total. It is difficult to ascertain a large number of reliably phenotyped postmortem brain tissues from autistic individuals given the scarcity of the resource32. This is a problem the field of ASD faces, generally. While this study focused on brain tissue, due to disease relevance and a lack of DNAm data in previous autism brain studies, there is also a need for complementary autism epigenomic studies in peripheral tissues, such as blood. Blood-based samples are particularly useful in ASD because large numbers of samples can be collected and because useful biomarkers for disease are needed and most likely will come from this tissue, as opposed to brain tissue, for practical reasons. Second, the DMR we identified at SDHAP3 in cerebellum showed a DNAm change in several autistic individuals that was potentially also related to a CNV. The 450K array is now the primary platform utilized for large epidemiology studies and this result highlights the need to examine and recognize the potential contribution of CNVs to methylation signals in these large studies. In addition, this finding demonstrates the need for new studies to clarify the relationships between CNVs and DNA methylation.

Despite these limitations, the findings presented in this study are of significance to the field of ASD for several reasons. To our knowledge, this is the first study to examine DNAm in ASD brains at genome-scale in regions outside of CpG islands and promoters, identifying 4 DMRs associated with the disorder. Second, while traditional genetic studies have identified rare variants in a minority of ASD cases, here, we identified genomic regions that commonly show differential methylation between autistic and control individuals. Lastly, the DMRs themselves are useful candidate regions for follow up studies.

Supplementary Material

Suppl Figure 1
Suppl Figure 2
Suppl Tables 1 & 2

Acknowledgments

We thank Daniel Geschwind and Neelroop Parikshak for sharing prefrontal and temporal cortex samples, obtained from the Autism Tissue Program (ATP) of Autism Speaks, for these analyses. In addition, we’d also like to thank the NICHD Brain and Tissue Bank for Neurodevelopmental Disorders at The University of Maryland for providing brain samples from the cerebellum brain region. This work was supported by the US National Institutes of Health Centers of Excellence in Genomic Science, 5P50HG003233 to A.P.F and Department of Defense (CDMRP) AR080125 to APF and WEK.

Footnotes

CONFLICT OF INTEREST

The authors declare no conflict of interest

Supplementary information is available at Molecular Psychiatry’s website

References

  • 1.Rapin I. Autism. N Engl J Med. 1997;337(2):97–104. doi: 10.1056/NEJM199707103370206. [DOI] [PubMed] [Google Scholar]
  • 2.American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 4. American Psychiatric Association Press; 1994. [Google Scholar]
  • 3.Autism: economic impact and implications. Proceedings of the 2012 Autism Summit; March 31, 2012; Hong Kong, China. 2012. [Google Scholar]
  • 4.Dawson G. Dramatic Increase in Autism Prevalence Parallels Explosion of Research Into Its Biology and Causes. Arch Gen Psychiatry. 2012:1–2. doi: 10.1001/jamapsychiatry.2013.488. [DOI] [PubMed] [Google Scholar]
  • 5.Splawski I, Yoo DS, Stotz SC, Cherry A, Clapham DE, Keating MT. CACNA1H mutations in autism spectrum disorders. J Biol Chem. 2006;281(31):22085–22091. doi: 10.1074/jbc.M603316200. [DOI] [PubMed] [Google Scholar]
  • 6.Heron SE, Khosravani H, Varela D, Bladen C, Williams TC, Newman MR, et al. Extended spectrum of idiopathic generalized epilepsies associated with CACNA1H functional variants. Ann Neurol. 2007;62(6):560–568. doi: 10.1002/ana.21169. [DOI] [PubMed] [Google Scholar]
  • 7.Glessner JT, Wang K, Cai G, Korvatska O, Kim CE, Wood S, et al. Autism genome-wide copy number variation reveals ubiquitin and neuronal genes. Nature. 2009;459(7246):569–573. doi: 10.1038/nature07953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cottrell CE, Bir N, Varga E, Alvarez CE, Bouyain S, Zernzach R, et al. Contactin 4 as an autism susceptibility locus. Autism Res. 2011;4(3):189–199. doi: 10.1002/aur.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Roohi J, Montagna C, Tegay DH, Palmer LE, DeVincent C, Pomeroy JC, et al. Disruption of contactin 4 in three subjects with autism spectrum disorder. J Med Genet. 2009;46(3):176–182. doi: 10.1136/jmg.2008.057505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Fernandez T, Morgan T, Davis N, Klin A, Morris A, Farhi A, et al. Disruption of Contactin 4 (CNTN4) results in developmental delay and other features of 3p deletion syndrome. Am J Hum Genet. 2008;82(6):1385. doi: 10.1016/j.ajhg.2008.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Banerjee-Basu S, Packer A. SFARI Gene: an evolving database for the autism research community. Dis Model Mech. 2010;3(3–4):133–135. doi: 10.1242/dmm.005439. [DOI] [PubMed] [Google Scholar]
  • 12.Newschaffer CJ, Fallin D, Lee NL. Heritable and nonheritable risk factors for autism spectrum disorders. Epidemiol Rev. 2002;24(2):137–153. doi: 10.1093/epirev/mxf010. [DOI] [PubMed] [Google Scholar]
  • 13.Sullivan PF, Daly MJ, O’Donovan M. Genetic architectures of psychiatric disorders: the emerging picture and its implications. Nat Rev Genet. 2012;13(8):537–551. doi: 10.1038/nrg3240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Schanen NC. Epigenetics of autism spectrum disorders. Hum Mol Genet. 2006;15(Spec No 2):R138–150. doi: 10.1093/hmg/ddl213. [DOI] [PubMed] [Google Scholar]
  • 15.Arking DE, Cutler DJ, Brune CW, Teslovich TM, West K, Ikeda M, et al. A common genetic variant in the neurexin superfamily member CNTNAP2 increases familial risk of autism. Am J Hum Genet. 2008;82(1):160–164. doi: 10.1016/j.ajhg.2007.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fradin D, Cheslack-Postava K, Ladd-Acosta C, Newschaffer C, Chakravarti A, Arking DE, et al. Parent-of-origin effects in autism identified through genome-wide linkage analysis of 16,000 SNPs. PLoS One. 2010;5(9) doi: 10.1371/journal.pone.0012513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet. 1999;23(2):185–188. doi: 10.1038/13810. [DOI] [PubMed] [Google Scholar]
  • 18.Horsthemke B, Wagstaff J. Mechanisms of imprinting of the Prader-Willi/Angelman region. Am J Med Genet A. 2008;146A(16):2041–2052. doi: 10.1002/ajmg.a.32364. [DOI] [PubMed] [Google Scholar]
  • 19.Oberle I, Rousseau F, Heitz D, Kretz C, Devys D, Hanauer A, et al. Instability of a 550-base pair DNA segment and abnormal methylation in fragile X syndrome. Science. 1991;252(5009):1097–1102. doi: 10.1126/science.252.5009.1097. [DOI] [PubMed] [Google Scholar]
  • 20.Ginsberg MR, Rubin RA, Falcone T, Ting AH, Natowicz MR. Brain transcriptional and epigenetic associations with autism. PLoS One. 2012;7(9):e44736. doi: 10.1371/journal.pone.0044736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nguyen A, Rauch TA, Pfeifer GP, Hu VW. Global methylation profiling of lymphoblastoid cell lines reveals epigenetic contributions to autism spectrum disorders and a novel autism candidate gene, RORA, whose protein product is reduced in autistic brain. Faseb J. 2010;24(8):3036–3051. doi: 10.1096/fj.10-154484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Irizarry RA, Ladd-Acosta C, Wen B, Wu Z, Montano C, Onyango P, et al. The human colon cancer methylome shows similar hypo- and hypermethylation at conserved tissue-specific CpG island shores. Nat Genet. 2009;41(2):178–186. doi: 10.1038/ng.298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Doi A, Park IH, Wen B, Murakami P, Aryee MJ, Irizarry R, et al. Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet. 2009;41(12):1350–1353. doi: 10.1038/ng.471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dudziec E, Miah S, Choudhry HM, Owen HC, Blizard S, Glover M, et al. Hypermethylation of CpG islands and shores around specific microRNAs and mirtrons is associated with the phenotype and presence of bladder cancer. Clin Cancer Res. 2011;17(6):1287–1296. doi: 10.1158/1078-0432.CCR-10-2017. [DOI] [PubMed] [Google Scholar]
  • 25.Feber A, Wilson GA, Zhang L, Presneau N, Idowu B, Down TA, et al. Comparative methylome analysis of benign and malignant peripheral nerve sheath tumors. Genome Res. 2011;21(4):515–524. doi: 10.1101/gr.109678.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rao X, Evans J, Chae H, Pilrose J, Kim S, Yan P, et al. CpG island shore methylation regulates caveolin-1 expression in breast cancer. Oncogene. 2012 doi: 10.1038/onc.2012.474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Akalin A, Garrett-Bakelman FE, Kormaksson M, Busuttil J, Zhang L, Khrebtukova I, et al. Base-pair resolution DNA methylation sequencing reveals profoundly divergent epigenetic landscapes in acute myeloid leukemia. PLoS Genet. 2012;8(6):e1002781. doi: 10.1371/journal.pgen.1002781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Shen J, Wang S, Zhang YJ, Wu HC, Kibriya MG, Jasmine F, et al. Exploring genome-wide DNA methylation profiles altered in hepatocellular carcinoma using Infinium HumanMethylation 450 BeadChips. Epigenetics. 2013;8(1):34–43. doi: 10.4161/epi.23062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Liu Y, Aryee MJ, Padyukov L, Fallin MD, Hesselberg E, Runarsson A, et al. Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in rheumatoid arthritis. Nat Biotechnol. 2013;31(2):142–147. doi: 10.1038/nbt.2487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Nakano K, Whitaker JW, Boyle DL, Wang W, Firestein GS. DNA methylome signature in rheumatoid arthritis. Ann Rheum Dis. 2013;72(1):110–117. doi: 10.1136/annrheumdis-2012-201526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Shulha HP, Cheung I, Whittle C, Wang J, Virgil D, Lin CL, et al. Epigenetic signatures of autism: trimethylated H3K4 landscapes in prefrontal neurons. Arch Gen Psychiatry. 2012;69(3):314–324. doi: 10.1001/archgenpsychiatry.2011.151. [DOI] [PubMed] [Google Scholar]
  • 32.Carey B. The New York Times. 2012. Brain Banks For Autism Face Dearth. [Google Scholar]
  • 33.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):380–384. doi: 10.1038/nature10110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bibikova M, Barnes B, Tsan C, Ho V, Klotzle B, Le JM, et al. High density DNA methylation array with single CpG site resolution. Genomics. 2011;98(4):288–295. doi: 10.1016/j.ygeno.2011.07.007. [DOI] [PubMed] [Google Scholar]
  • 35.Sandoval J, Heyn H, Moran S, Serra-Musach J, Pujana MA, Bibikova M, et al. Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome. Epigenetics. 2011;6(6):692–702. doi: 10.4161/epi.6.6.16196. [DOI] [PubMed] [Google Scholar]
  • 36.Jaffe AE, Murakami P, Lee H, Leek JT, Fallin MD, Feinberg AP, et al. Bump hunting to identify differentially methylated regions in epigenetic epidemiology studies. Int J Epidemiol. 2012;41(1):200–209. doi: 10.1093/ije/dyr238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hansen KAM, Irizarry RA. Minfi: analyzing Illumina 450K methylation arrays. 2012. [Google Scholar]
  • 38.Du P, Zhang X, Huang CC, Jafari N, Kibbe WA, Hou L, et al. Comparison of Beta-value and M-value methods for quantifying methylation levels by microarray analysis. BMC Bioinformatics. 2010;11:587. doi: 10.1186/1471-2105-11-587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Guintivano J, Aryee M, Kaminsky Z. A cell epigenotype specific model for the correction of brain cellular heterogeneity bias and its application to age, brain region and major depression. Epigenetics. 2013;8(3) doi: 10.4161/epi.23924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Krek A, Grun D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, et al. Combinatorial microRNA target predictions. Nat Genet. 2005;37(5):495–500. doi: 10.1038/ng1536. [DOI] [PubMed] [Google Scholar]
  • 41.Lai EC. Micro RNAs are complementary to 3′ UTR sequence motifs that mediate negative post-transcriptional regulation. Nat Genet. 2002;30(4):363–364. doi: 10.1038/ng865. [DOI] [PubMed] [Google Scholar]
  • 42.Fukuoka T, Sumida K, Yamada T, Higuchi C, Nakagaki K, Nakamura K, et al. Gene expression profiles in the common marmoset brain determined using a newly developed common marmoset-specific DNA microarray. Neurosci Res. 2010;66(1):62–85. doi: 10.1016/j.neures.2009.09.1709. [DOI] [PubMed] [Google Scholar]
  • 43.Groen W, Teluij M, Buitelaar J, Tendolkar I. Amygdala and hippocampus enlargement during adolescence in autism. J Am Acad Child Adolesc Psychiatry. 2010;49(6):552–560. doi: 10.1016/j.jaac.2009.12.023. [DOI] [PubMed] [Google Scholar]
  • 44.Hasan KM, Walimuni IS, Frye RE. Global Cerebral and Regional Multimodal Neuroimaging Markers of the Neurobiology of Autism: Development and Cognition. J Child Neurol. 2012 doi: 10.1177/0883073812452917. [DOI] [PubMed] [Google Scholar]
  • 45.Agostini G, Mancini J, Chabrol B, Villeneuve N, Milh M, George F, et al. Language disorders in children with morphologic abnormalities of the hippocampus. Arch Pediatr. 2010;17(7):1008–1016. doi: 10.1016/j.arcped.2010.03.002. [DOI] [PubMed] [Google Scholar]
  • 46.Raymond GV, Bauman ML, Kemper TL. Hippocampus in autism: a Golgi analysis. Acta neuropathologica. 1996;91(1):117–119. doi: 10.1007/s004010050401. [DOI] [PubMed] [Google Scholar]
  • 47.Clement JP, Aceti M, Creson TK, Ozkan ED, Shi Y, Reish NJ, et al. Pathogenic SYNGAP1 mutations impair cognitive development by disrupting maturation of dendritic spine synapses. Cell. 2012;151(4):709–723. doi: 10.1016/j.cell.2012.08.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Labate A, Tarantino P, Palamara G, Gagliardi M, Cavalcanti F, Ferlazzo E, et al. Mutations in PRRT2 result in familial infantile seizures with heterogeneous phenotypes including febrile convulsions and probable SUDEP. Epilepsy Res. 2013 doi: 10.1016/j.eplepsyres.2012.10.014. [DOI] [PubMed] [Google Scholar]
  • 49.Chen WJ, Lin Y, Xiong ZQ, Wei W, Ni W, Tan GH, et al. Exome sequencing identifies truncating mutations in PRRT2 that cause paroxysmal kinesigenic dyskinesia. Nat Genet. 2011;43(12):1252–1255. doi: 10.1038/ng.1008. [DOI] [PubMed] [Google Scholar]
  • 50.Riant F, Roze E, Barbance C, Meneret A, Guyant-Marechal L, Lucas C, et al. PRRT2 mutations cause hemiplegic migraine. Neurology. 2012;79(21):2122–2124. doi: 10.1212/WNL.0b013e3182752cb8. [DOI] [PubMed] [Google Scholar]
  • 51.Lee HY, Huang Y, Bruneau N, Roll P, Roberson ED, Hermann M, et al. Mutations in the gene PRRT2 cause paroxysmal kinesigenic dyskinesia with infantile convulsions. Cell Rep. 2012;1(1):2–12. doi: 10.1016/j.celrep.2011.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Heron SE, Grinton BE, Kivity S, Afawi Z, Zuberi SM, Hughes JN, et al. PRRT2 mutations cause benign familial infantile epilepsy and infantile convulsions with choreoathetosis syndrome. Am J Hum Genet. 2012;90(1):152–160. doi: 10.1016/j.ajhg.2011.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.de Vries B, Callenbach PM, Kamphorst JT, Weller CM, Koelewijn SC, ten Houten R, et al. PRRT2 mutation causes benign familial infantile convulsions. Neurology. 2012;79(21):2154–2155. doi: 10.1212/WNL.0b013e3182752c30. [DOI] [PubMed] [Google Scholar]
  • 54.Ono S, Yoshiura K, Kinoshita A, Kikuchi T, Nakane Y, Kato N, et al. Mutations in PRRT2 responsible for paroxysmal kinesigenic dyskinesias also cause benign familial infantile convulsions. J Hum Genet. 2012;57(5):338–341. doi: 10.1038/jhg.2012.23. [DOI] [PubMed] [Google Scholar]
  • 55.Gartlan KH, Belz GT, Tarrant JM, Minigo G, Katsara M, Sheng KC, et al. A complementary role for the tetraspanins CD37 and Tssc6 in cellular immunity. J Immunol. 2010;185(6):3158–3166. doi: 10.4049/jimmunol.0902867. [DOI] [PubMed] [Google Scholar]
  • 56.Goschnick MW, Jackson DE. Tetraspanins-structural and signalling scaffolds that regulate platelet function. Mini Rev Med Chem. 2007;7(12):1248–1254. doi: 10.2174/138955707782795656. [DOI] [PubMed] [Google Scholar]
  • 57.Scholz CJ, Jacob CP, Buttenschon HN, Kittel-Schneider S, Boreatti-Hummer A, Zimmer M, et al. Functional variants of TSPAN8 are associated with bipolar disorder and schizophrenia. Am J Med Genet B Neuropsychiatr Genet. 2010;153B(4):967–972. doi: 10.1002/ajmg.b.31057. [DOI] [PubMed] [Google Scholar]
  • 58.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(1):25–27. doi: 10.1038/ng1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Moessner R, Marshall CR, Sutcliffe JS, Skaug J, Pinto D, Vincent J, et al. Contribution of SHANK3 mutations to autism spectrum disorder. Am J Hum Genet. 2007;81(6):1289–1297. doi: 10.1086/522590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Gauthier J, Spiegelman D, Piton A, Lafreniere RG, Laurent S, St-Onge J, et al. Novel de novo SHANK3 mutation in autistic patients. Am J Med Genet B Neuropsychiatr Genet. 2009;150B(3):421–424. doi: 10.1002/ajmg.b.30822. [DOI] [PubMed] [Google Scholar]
  • 61.Berkel S, Marshall CR, Weiss B, Howe J, Roeth R, Moog U, et al. Mutations in the SHANK2 synaptic scaffolding gene in autism spectrum disorder and mental retardation. Nat Genet. 2010;42(6):489–491. doi: 10.1038/ng.589. [DOI] [PubMed] [Google Scholar]
  • 62.Volders K, Nuytens K, Creemers JW. The autism candidate gene Neurobeachin encodes a scaffolding protein implicated in membrane trafficking and signaling. Curr Mol Med. 2011;11(3):204–217. doi: 10.2174/156652411795243432. [DOI] [PubMed] [Google Scholar]
  • 63.Hogart A, Wu D, LaSalle JM, Schanen NC. The comorbidity of autism with the genomic disorders of chromosome 15q11. 2-q13. Neurobiol Dis. 2010;38(2):181–191. doi: 10.1016/j.nbd.2008.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Li X, Ito M, Zhou F, Youngson N, Zuo X, Leder P, et al. A maternal-zygotic effect gene, Zfp57, maintains both maternal and paternal imprints. Dev Cell. 2008;15(4):547–557. doi: 10.1016/j.devcel.2008.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Strogantsev R, Ferguson-Smith AC. Proteins involved in establishment and maintenance of imprinted methylation marks. Brief Funct Genomics. 2012;11(3):227–239. doi: 10.1093/bfgp/els018. [DOI] [PubMed] [Google Scholar]
  • 66.Zuo X, Sheng J, Lau HT, McDonald CM, Andrade M, Cullen DE, et al. Zinc finger protein ZFP57 requires its co-factor to recruit DNA methyltransferases and maintains DNA methylation imprint in embryonic stem cells via its transcriptional repression domain. J Biol Chem. 2012;287(3):2107–2118. doi: 10.1074/jbc.M111.322644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Liu Y, Zhang X, Blumenthal RM, Cheng X. A common mode of recognition for methylated CpG. Trends Biochem Sci. 2013 doi: 10.1016/j.tibs.2012.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Raney BJ, Cline MS, Rosenbloom KR, Dreszer TR, Learned K, Barber GP, et al. ENCODE whole-genome data in the UCSC genome browser (2011 update) Nucleic Acids Res. 2011;39(Database issue):D871–875. doi: 10.1093/nar/gkq1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Rossignol DA, Frye RE. Mitochondrial dysfunction in autism spectrum disorders: a systematic review and meta-analysis. Mol Psychiatry. 2012;17(3):290–314. doi: 10.1038/mp.2010.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Marui T, Funatogawa I, Koishi S, Yamamoto K, Matsumoto H, Hashimoto O, et al. The NADH-ubiquinone oxidoreductase 1 alpha subcomplex 5 (NDUFA5) gene variants are associated with autism. Acta Psychiatr Scand. 2011;123(2):118–124. doi: 10.1111/j.1600-0447.2010.01600.x. [DOI] [PubMed] [Google Scholar]
  • 71.Tsao CY, Mendell JR. Autistic disorder in 2 children with mitochondrial disorders. J Child Neurol. 2007;22(9):1121–1123. doi: 10.1177/0883073807306266. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Suppl Figure 1
Suppl Figure 2
Suppl Tables 1 & 2

RESOURCES