Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex

Abstract

DNA methylation (DNAm) is important in brain development and is potentially important in schizophrenia. We characterized DNAm in prefrontal cortex from 335 non-psychiatric controls across the lifespan and 191 patients with schizophrenia and identified widespread changes in the transition from prenatal to postnatal life. These DNAm changes manifest in the transcriptome, correlate strongly with a shifting cellular landscape and overlap regions of genetic risk for schizophrenia. A quarter of published genome-wide association studies (GWAS)-suggestive loci (4,208 of 15,930, P < 10−100) manifest as significant methylation quantitative trait loci (meQTLs), including 59.6% of GWAS-positive schizophrenia loci. We identified 2,104 CpGs that differ between schizophrenia patients and controls that were enriched for genes related to development and neurodifferentiation. The schizophrenia-associated CpGs strongly correlate with changes related to the prenatal-postnatal transition and show slight enrichment for GWAS risk loci while not corresponding to CpGs differentiating adolescence from later adult life. These data implicate an epigenetic component to the developmental origins of this disorder.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Differentially methylated loci comparing pre- and postnatal control subjects show large differences in DNA methylation.
Figure 2: A changing neuronal phenotype across brain development.
Figure 3: Examples of meQTLs for six GWAS-associated variants with nearby DNA methylation levels.
Figure 4: Examples of meQTLs for 12 GWAS-positive loci for schizophrenia.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Colantuoni, C. et al. Temporal dynamics and genetic control of transcription in the human prefrontal cortex. Nature 478, 519–523 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Numata, S. et al. DNA methylation signatures in development and aging of the human prefrontal cortex. Am. J. Hum. Genet. 90, 260–272 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Grayson, D.R. & Guidotti, A. The dynamics of DNA methylation in schizophrenia and related psychiatric disorders. Neuropsychopharmacology 38, 138–166 (2013).

    CAS  PubMed  Google Scholar 

  4. Waterland, R.A. & Michels, K.B. Epigenetic epidemiology of the developmental origins hypothesis. Annu. Rev. Nutr. 27, 363–388 (2007).

    CAS  PubMed  Google Scholar 

  5. Jakovcevski, M. & Akbarian, S. Epigenetic mechanisms in neurological disease. Nat. Med. 18, 1194–1204 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Weinberger, D.R. & Levitt, P. Neurodevelopmental origins of schizophrenia. in Schizophrenia (eds. Weinberger, D.R. & Harrison, P.J.) (Wiley-Blackwell, 2011).

  7. Heijmans, B.T. et al. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc. Natl. Acad. Sci. USA 105, 17046–17049 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Breitling, L.P., Yang, R., Korn, B., Burwinkel, B. & Brenner, H. Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am. J. Hum. Genet. 88, 450–457 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Reichard, J.F., Schnekenburger, M. & Puga, A. Long term low-dose arsenic exposure induces loss of DNA methylation. Biochem. Biophys. Res. Commun. 352, 188–192 (2007).

    CAS  PubMed  Google Scholar 

  10. Susser, E.S. & Lin, S.P. Schizophrenia after prenatal exposure to the Dutch Hunger Winter of 1944–1945. Arch. Gen. Psychiatry 49, 983–988 (1992).

    CAS  PubMed  Google Scholar 

  11. Relton, C.L. & Davey Smith, G. Is epidemiology ready for epigenetics? Int. J. Epidemiol. 41, 5–9 (2012).

    PubMed  PubMed Central  Google Scholar 

  12. Cortessis, V.K. et al. Environmental epigenetics: prospects for studying epigenetic mediation of exposure-response relationships. Hum. Genet. 131, 1565–1589 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Schilling, E., El Chartouni, C. & Rehli, M. Allele-specific DNA methylation in mouse strains is mainly determined by cis-acting sequences. Genome Res. 19, 2028–2035 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Lienert, F. et al. Identification of genetic elements that autonomously determine DNA methylation states. Nat. Genet. 43, 1091–1097 (2011).

    CAS  PubMed  Google Scholar 

  15. Bird, A. Putting the DNA back into DNA methylation. Nat. Genet. 43, 1050–1051 (2011).

    CAS  PubMed  Google Scholar 

  16. Lister, R. et al. Global epigenomic reconfiguration during mammalian brain development. Science 341, 1237905 (2013).

    PubMed  PubMed Central  Google Scholar 

  17. Sandoval, J. et al. Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome. Epigenetics 6, 692–702 (2011).

    CAS  PubMed  Google Scholar 

  18. Jaffe, A.E. et al. Bump hunting to identify differentially methylated regions in epigenetic epidemiology studies. Int. J. Epidemiol. 41, 200–209 (2012).

    PubMed  PubMed Central  Google Scholar 

  19. Hansen, K.D. et al. Increased methylation variation in epigenetic domains across cancer types. Nat. Genet. 43, 768–775 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Aryee, M.J. et al. Minfi: a flexible and comprehensive Bioconductor package for the analysis of Infinium DNA methylation microarrays. Bioinformatics 30, 1363–1369 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Timp, W. et al. Large hypomethylated blocks as a universal defining epigenetic alteration in human solid tumors. Genome Med. 6, 61 (2014).

    PubMed  PubMed Central  Google Scholar 

  22. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Jaffe, A.E. et al. Developmental regulation of human cortex transcription and its clinical relevance at single base resolution. Nat. Neurosci. 18, 154–161 (2015).

    CAS  PubMed  Google Scholar 

  24. Spiers, H. et al. Methylomic trajectories across human fetal brain development. Genome Res. 25, 338–352 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. BrainSpan. Atlas of the Developing Human Brain, &lt;http://developinghumanbrain.org&gt; (2011).

  26. Schizophrenia Working Group of the Psychiatric Genomics. C. Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427 (2014).

  27. Lambert, J.C. et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat. Genet. 45, 1452–1458 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Nalls, M.A. et al. Imputation of sequence variants for identification of genetic risks for Parkinson′s disease: a meta-analysis of genome-wide association studies. Lancet 377, 641–649 (2011).

    PubMed  Google Scholar 

  29. Morris, A.P. et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat. Genet. 44, 981–990 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Hannon, E. et al. Methylation QTLs in the developing brain: enrichment in schizophrenia-associated genomic regions. Nat. Neurosci. advance online publication, doi:10.1038/nn.4182 (30 November 2015).

  31. Liu, Y. et al. Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in rheumatoid arthritis. Nat. Biotechnol. 31, 142–147 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Welter, D. et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Res. 42, D1001–D1006 (2014).

    CAS  PubMed  Google Scholar 

  33. Nichols, L. et al. The National Institutes of Health Neurobiobank: a federated national network of human brain and tissue repositories. Biol. Psychiatry 75, e21–e22 (2014).

    PubMed  Google Scholar 

  34. Falcon, S. & Gentleman, R. Using GOstats to test gene lists for GO term association. Bioinformatics 23, 257–258 (2007).

    CAS  PubMed  Google Scholar 

  35. Verreault, A., Kaufman, P.D., Kobayashi, R. & Stillman, B. Nucleosomal DNA regulates the core-histone-binding subunit of the human Hat1 acetyltransferase. Curr. Biol. 8, 96–108 (1998).

    CAS  PubMed  Google Scholar 

  36. Gavin, D.P. & Sharma, R.P. Histone modifications, DNA methylation and schizophrenia. Neurosci. Biobehav. Rev. 34, 882–888 (2010).

    CAS  PubMed  Google Scholar 

  37. Pidsley, R. et al. Methylomic profiling of human brain tissue supports a neurodevelopmental origin for schizophrenia. Genome Biol. 15, 483 (2014).

    PubMed  PubMed Central  Google Scholar 

  38. Darmanis, S. et al. A survey of human brain transcriptome diversity at the single cell level. Proc. Natl. Acad. Sci. USA 112, 7285–7290 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Abdolmaleky, H.M. et al. Methylomics in psychiatry: Modulation of gene-environment interactions may be through DNA methylation. Am. J. Med. Genet. B Neuropsychiatr. Genet. 127B, 51–59 (2004).

    PubMed  Google Scholar 

  40. Mill, J. et al. Epigenomic profiling reveals DNA-methylation changes associated with major psychosis. Am. J. Hum. Genet. 82, 696–711 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Gulsuner, S. et al. Spatial and temporal mapping of de novo mutations in schizophrenia to a fetal prefrontal cortical network. Cell 154, 518–529 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Parikshak, N.N. et al. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell 155, 1008–1021 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Kleinman, J.E. et al. Genetic neuropathology of schizophrenia: new approaches to an old question and new uses for postmortem human brains. Biol. Psychiatry 69, 140–145 (2011).

    PubMed  PubMed Central  Google Scholar 

  44. The PsychENCODE Consortium. The PsychENCODE Project. Nat. Neurosci. 18, 1708–1713 (2015).

  45. Lipska, B.K. et al. Critical factors in gene expression in postmortem human brain: focus on studies in schizophrenia. Biol. Psychiatry 60, 650–658 (2006).

    CAS  PubMed  Google Scholar 

  46. Deep-Soboslay, A. et al. Reliability of psychiatric diagnosis in postmortem research. Biol. Psychiatry 57, 96–101 (2005).

    PubMed  Google Scholar 

  47. Smyth, G.K. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3, 3 (2004).

    Google Scholar 

  48. Bland, J.M. & Altman, D.G. Multiple significance tests: the Bonferroni method. Br. Med. J. 310, 170 (1995).

    CAS  Google Scholar 

  49. Irizarry, R.A. et al. Comprehensive high-throughput arrays for relative methylation (CHARM). Genome Res. 18, 780–790 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Farrell, C.M. . et al. Current status and new features of the Consensus Coding Sequence database. Nucleic Acids Res. 42, D865–D872 (2014).

    CAS  PubMed  Google Scholar 

  51. Zoubarev, A. et al. Gemma: a resource for the reuse, sharing and meta-analysis of expression profiling data. Bioinformatics 28, 2272–2273 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Kim, M. et al. Dynamic changes in DNA methylation and hydroxymethylation when hES cells undergo differentiation toward a neuronal lineage. Hum. Mol. Genet. 23, 657–667 (2014).

    CAS  PubMed  Google Scholar 

  53. Guintivano, J., Aryee, M.J. & Kaminsky, Z.A. A cell epigenotype specific model for the correction of brain cellular heterogeneity bias and its application to age, brain region and major depression. Epigenetics 8, 290–302 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Houseman, E.A. et al. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinformatics 13, 86 (2012).

    PubMed  PubMed Central  Google Scholar 

  55. Edgar, R., Domrachev, M. & Lash, A.E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30, 207–210 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Jaffe, A.E. & Irizarry, R.A. Accounting for cellular heterogeneity is critical in epigenome-wide association studies. Genome Biol. 15, R31 (2014).

    PubMed  PubMed Central  Google Scholar 

  57. Scharpf, R.B., Irizarry, R.A., Ritchie, M.E., Carvalho, B. & Ruczinski, I. Using the R Package crlmm for genotyping and copy number estimation. J. Stat. Softw. 40, 1–32 (2011).

    PubMed  PubMed Central  Google Scholar 

  58. Anderson, C.A. et al. Data quality control in genetic case-control association studies. Nat. Protoc. 5, 1564–1573 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Williams, A.L., Patterson, N., Glessner, J., Hakonarson, H. & Reich, D. Phasing of many thousands of genotyped samples. Am. J. Hum. Genet. 91, 238–251 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Howie, B.N., Donnelly, P. & Marchini, J. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet. 5, e1000529 (2009).

    PubMed  PubMed Central  Google Scholar 

  61. Shabalin, A.A. Matrix eQTL: ultra fast eQTL analysis via large matrix operations. Bioinformatics 28, 1353–1358 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful for the vision and generosity of the Lieber and Maltz Families who made this work possible. We thank the families who donated to this research and we thank A. Feinberg for helpful input on data analyses. This work was supported by the Lieber Institute for Brain Development. A.E.J. was partially supported by 1R21MH102791.

Author information

Authors and Affiliations

Authors

Contributions

A.E.J. designed the study, performed the data analysis and oversaw the writing of the manuscript. Y.G. oversaw the data generation. A.D.-S. collected phenotype data on all subjects. R.T. performed DNA extractions and contributed to the data generation. T.M.H. collected brain samples and performed tissue dissections to obtain biological materials. D.R.W. designed the study, contributed to the data analysis and interpretation of the results, and oversaw the writing of the manuscript. J.E.K. collected brain samples and provided clinical interpretation of the results. All authors contributed to the writing of the manuscript.

Corresponding author

Correspondence to Andrew E Jaffe.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Principal component analysis (PCA) demonstrates genome-wide changes in DNAm comparing pre- and post-natal samples.

The first principal component (PC, explaining 55.6% of the variance in the data) cleanly separately pre- and post-natal samples, with samples from children (ages 0-13) demonstrating directional consistency across aging.

Supplementary Figure 2 Example differentially methylated regions (DMRs) for pre- versus post-natal differences in DNA methylation, as Figure 1B.

Proportion methylation is shown on the y-axis of each respective top-most panel. Gene annotation panels in (B) and (C) are based on Ensembl annotation – dark blue represents exons and light blue represents introns.

Supplementary Figure 3 Directionally balanced associations between DNAm and nearby gene expression levels at individual CpGs.

Pearson correlations between DNAm levels at individual CpGs/probes and nearby genes, which contains approximately equal numbers of positive and negative correlations.

Supplementary Figure 4 Directionally balanced associations between DNAm and nearby gene expression levels at DMRs.

Pearson correlations between DNAm levels at DMRs to nearby genes, stratified by the location of the DMR relative to that gene. These correlations are relatively balanced for positive and negative correlations overall, and by many classes of annotation.

Supplementary Figure 5 Proportion of variance explained in gene expression levels by cell composition estimated from DNAm data.

Supplementary Figure 6 Composition change across fetal brain development.

Shown are the composition estimates versus post-conception data for the 4 additional cell types, with Pearson correlation printed in the top right corner, as Figure 2F

Supplementary Figure 7 Neuronal composition differences by brain region and age in the BrainSpan dataset.

Each point is a sample, colored by age, and stratified by brain regions. The vertical dashed line separates the cortical brain regions from non-cortical regions.

Supplementary Figure 8 Effect of adult meQTLs in fetal samples.

Supplementary Figure 9 Cellular composition profiles by diagnosis.

(A) Distributions of NeuN- estimates per sample by processing plate and diagnosis; all p-values for diagnosis within a plate were > 0.01. (B) NeuN- proportion explains the first principal component of autosomal DNAm levels in adult samples.

Supplementary Figure 10 Negative control principal components associate with processing plate and slide.

Supplementary Figure 11 Samples on one processing plate (“Plate2”) show magnified differential methylation effects for schizophrenia.

While the T-statistics within each plate are correlated, those calculated only within samples on Plate2 were almost an order of magnitude larger than similar statistics calculated only within samples on Plate3. Red: identity line.

Supplementary Figure 12 Sensitivity analyses of differentially methylated CpGs for schizophrenia.

We compared the effects sizes of schizophrenia differences from the original statistical model (adjusting for age, sex, race, and 4 negative control PCs) to also including (A) composition estimates from all 5 cell types and (B) smoking status determined by toxicology and (C) antipsychotics by self-report in the final model.

Supplementary Figure 13 Effect of adult control meQTLs in adult SZ samples.

Supplementary Figure 14 Concordance between schizophrenia and developmental effects across 2,104 CpGs associated with schizophrenia.

X-axis: change in the DNA methylation (DNAm) levels comparing prenatal versus postnatal samples, Y-axis: change in DNAm level comparing patients with schizophrenia to adult controls. Each point is one CpG probe.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–14 and Supplementary Analysis (PDF 6627 kb)

Supplementary Methods Checklist (PDF 384 kb)

Supplementary Table 1

Demographic data for the samples analyzed, stratified by age group and diagnosis. P-values depict the differences between the demographic data by the cases and adult controls. (XLSX 10 kb)

Supplementary Table 2

Differentially methylated regions (DMRs) and corresponding annotation comparing prenatal and postnatal samples. (CSV 1893 kb)

Supplementary Table 3

Gene ontology (GO) enrichment statistics for those DMRs that increase/“up” or decrease/“down” across the transition from pre to postnatal life (XLSX 933 kb)

Supplementary Table 4

Differentially methylated blocks comparing prenatal and postnatal samples (CSV 110 kb)

Supplementary Table 5

Gene ontology (GO) analysis on the genes contained with the differentially methylated blocks. (CSV 526 kb)

Supplementary Table 6

Overlap between DNAm changes and chromatin state data from the Epigenome Roadmap project for adult DLPFC Bolded cells indicated >2 fold enrichment or depletion compared to the relevant background CpGs/regions (XLSX 13 kb)

Supplementary Table 7

Overlap between PGC2 risk regions and DMRs associated with the transition from prenatal to postnatal life, n=31 regions. Rank: PGC Rank, P.value: p-value for the region, Position: range of LD block for region, numDMRs: the number of DMRs within the region. (CSV 4 kb)

Supplementary Table 8

NHGRI GWAS catalog annotated by whether each SNP has an meQTL in the DLPFC dataset. (XLSX 6171 kb)

Supplementary Table 9

meQTLs within the PGC2 SNPs and their proxies (XLSX 1465 kb)

Supplementary Table 10

List of differentially methylated CpGs comparing patients with schizophrenia to adult controls. (CSV 543 kb)

Supplementary Table 11

Gene ontology (GO) analysis for genes near differentially methylated CpGs for diagnosis. (CSV 883 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jaffe, A., Gao, Y., Deep-Soboslay, A. et al. Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex. Nat Neurosci 19, 40–47 (2016). https://doi.org/10.1038/nn.4181

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4181

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing