11 research outputs found

    The Promise of DNA Methylation in Understanding Multigenerational Factors in Autism Spectrum Disorders

    No full text
    Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders characterized by impairments in social reciprocity and communication, restrictive interests, and repetitive behaviors. Most cases of ASD arise from a confluence of genetic susceptibility and environmental risk factors, whose interactions can be studied through epigenetic mechanisms such as DNA methylation. While various parental factors are known to increase risk for ASD, several studies have indicated that grandparental and great-grandparental factors may also contribute. In animal studies, gestational exposure to certain environmental factors, such as insecticides, medications, and social stress, increases risk for altered behavioral phenotypes in multiple subsequent generations. Changes in DNA methylation, gene expression, and chromatin accessibility often accompany these altered behavioral phenotypes, with changes often appearing in genes that are important for neurodevelopment or have been previously implicated in ASD. One hypothesized mechanism for these phenotypic and methylation changes includes the transmission of DNA methylation marks at individual chromosomal loci from parent to offspring and beyond, called multigenerational epigenetic inheritance. Alternatively, intermediate metabolic phenotypes in the parental generation may confer risk from the original grandparental exposure to risk for ASD in grandchildren, mediated by DNA methylation. While hypothesized mechanisms require further research, the potential for multigenerational epigenetics assessments of ASD risk has implications for precision medicine as the field attempts to address the variable etiology and clinical signs of ASD by incorporating genetic, environmental, and lifestyle factors. In this review, we discuss the promise of multigenerational DNA methylation investigations in understanding the complex etiology of ASD

    Comethyl: a network-based methylome approach to investigate the multivariate nature of health and disease.

    No full text
    Health outcomes are frequently shaped by difficult to dissect inter-relationships between biological, behavioral, social and environmental factors. DNA methylation patterns reflect such multivariate intersections, providing a rich source of novel biomarkers and insight into disease etiologies. Recent advances in whole-genome bisulfite sequencing enable investigation of DNA methylation over all genomic CpGs, but existing bioinformatic approaches lack accessible system-level tools. Here, we develop the R package Comethyl, for weighted gene correlation network analysis of user-defined genomic regions that generates modules of comethylated regions, which are then tested for correlations with multivariate sample traits. First, regions are defined by CpG genomic location or regulatory annotation and filtered based on CpG count, sequencing depth and variability. Next, correlation networks are used to find modules of interconnected nodes using methylation values within the selected regions. Each module containing multiple comethylated regions is reduced in complexity to a single eigennode value, which is then tested for correlations with experimental metadata. Comethyl has the ability to cover the noncoding regulatory regions of the genome with high relevance to interpretation of genome-wide association studies and integration with other types of epigenomic data. We demonstrate the utility of Comethyl on a dataset of male cord blood samples from newborns later diagnosed with autism spectrum disorder (ASD) versus typical development. Comethyl successfully identified an ASD-associated module containing regions mapped to genes enriched for brain glial functions. Comethyl is expected to be useful in uncovering the multivariate nature of health disparities for a variety of common disorders. Comethyl is available at github.com/cemordaunt/comethyl with complete documentation and example analyses

    Epigenomic signature of major congenital heart defects in newborns with Down syndrome

    No full text
    Abstract Background Congenital heart defects (CHDs) affect approximately half of individuals with Down syndrome (DS), but the molecular reasons for incomplete penetrance are unknown. Previous studies have largely focused on identifying genetic risk factors associated with CHDs in individuals with DS, but comprehensive studies of the contribution of epigenetic marks are lacking. We aimed to identify and characterize DNA methylation differences from newborn dried blood spots (NDBS) of DS individuals with major CHDs compared to DS individuals without CHDs. Methods We used the Illumina EPIC array and whole-genome bisulfite sequencing (WGBS) to quantitate DNA methylation for 86 NDBS samples from the California Biobank Program: (1) 45 DS-CHD (27 female, 18 male) and (2) 41 DS non-CHD (27 female, 14 male). We analyzed global CpG methylation and identified differentially methylated regions (DMRs) in DS-CHD versus DS non-CHD comparisons (both sex-combined and sex-stratified) corrected for sex, age of blood collection, and cell-type proportions. CHD DMRs were analyzed for enrichment in CpG and genic contexts, chromatin states, and histone modifications by genomic coordinates and for gene ontology enrichment by gene mapping. DMRs were also tested in a replication dataset and compared to methylation levels in DS versus typical development (TD) WGBS NDBS samples. Results We found global CpG hypomethylation in DS-CHD males compared to DS non-CHD males, which was attributable to elevated levels of nucleated red blood cells and not seen in females. At a regional level, we identified 58, 341, and 3938 CHD-associated DMRs in the Sex Combined, Females Only, and Males Only groups, respectively, and used machine learning algorithms to select 19 Males Only loci that could distinguish CHD from non-CHD. DMRs in all comparisons were enriched for gene exons, CpG islands, and bivalent chromatin and mapped to genes enriched for terms related to cardiac and immune functions. Lastly, a greater percentage of CHD-associated DMRs than background regions were differentially methylated in DS versus TD samples. Conclusions A sex-specific signature of DNA methylation was detected in NDBS of DS-CHD compared to DS non-CHD individuals. This supports the hypothesis that epigenetics can reflect the variability of phenotypes in DS, particularly CHDs

    Additional file 1 of Epigenomic signature of major congenital heart defects in newborns with Down syndrome

    No full text
    Additional file 1: Table S1. Study characteristics: description of sample traits included in study. Table S2. Sample traits: values of traits for each sample. Table S3. Welch’s t test for differences in sample traits in DS-CHD versus DS non-CHD samples. Table S4. Pearson correlation coefficients and p values (unadjusted and adjusted) for sample traits. Table S5. EPIC array beta values. Table S6. Logistic regression for CHD and linear regression for global methylation. Table S7. Annotated Sex Combined DMRs (adjusted for sex, age of blood collection, cell types). Table S8. Annotated Females Only DMRs (adjusted for age of blood collection, cell types). Table S9. Annotated Males Only DMRs (adjusted for age of blood collection, cell types). Table S10. Annotated Males Only DMRs (adjusted for age of blood collection, cell types) Sensitivity Analysis (5 samples with nRBC > 20% removed). Table S11. Machine Learning DMRs. Table S12. Smoothed methylation of Sex-combined DMRs in replication dataset. Table S13. Smoothed methylation of Females Only DMRs in replication dataset. Table S14. Smoothed methylation of Males Only DMRs in replication dataset. Table S15. Stats from permutation testing of DMR overlaps from Sex Combined, Females Only, and Males Only comparisons. Table S16. Chromosome location enrichments of Sex Combined, Females Only, and Males Only DMRs. Table S17. CpG and Genic enrichments for Sex Combined, Females Only, and Males Only comparisons. Table S18. GREAT gene ontology enrichments for Sex-Combined DMRs compared to background regions. Table S19. GREAT gene ontology enrichments for Females Only DMRs compared to background regions. Table S20. GREAT gene ontology enrichments for Males Only DMRs compared to background regions. Table S21. Smoothed methylation of Sex-combined DMRs in DSvTD dataset. Table S22. Smoothed methylation of Females Only DMRs in DSvTD dataset. Table S23. Smoothed methylation of Males Only DMRs in DSvTD dataset
    corecore