Neuronal brain region-specific DNA methylation and chromatin accessibility are associated with neuropsychiatric disease heritability

Epigenetic modifications confer stable transcriptional patterns in the brain, and both normal and abnormal brain function involve specialized brain regions, yet little is known about brain region-specific epigenetic differences. Here, we compared prefrontal cortex, anterior cingulate gyrus, hippocampus and nucleus accumbens from 6 individuals, performing whole genome bisulfite sequencing for DNA methylation. In addition, we have performed ATAC-seq for chromatin accessibility, and RNA-seq for gene expression in the nucleus accumbens and prefrontal cortex from 6 additional individuals. We found substantial neuron- and brain region-specific differences in both DNA methylation and chromatin accessibility which were largely non-overlapping, and were greatest between nucleus accumbens and the other regions. In contrast, glial methylation and chromatin were relatively homogeneous across brain regions, although neuron/glia ratios varied greatly, demonstrating the necessity for cellular fractionation. Gene expression was also largely the same across glia from different brain regions and substantially different for neurons. Expression was correlated with methylation and accessibility across promoters and known enhancers. Several classes of transcription factor binding sites were enriched at regions of differential methylation and accessibility, including many that respond to synaptic activity. Finally, both regions of differential methylation and those of differential accessibility showed a surprising >10-fold enrichment of explained heritability associated with addictive behavior, as well as schizophrenia- and neuroticism-associated regions, suggesting that common psychiatric illness is mediated through brain region-specific epigenetic marks.

[1]  H. Maeno,et al.  Dopamine Receptors , 2018 .

[2]  M. Frommer,et al.  CpG islands in vertebrate genomes. , 1987, Journal of molecular biology.

[3]  J. McGinty,et al.  A single injection of amphetamine or methamphetamine induces dynamic alterations in c-fos,zif/268 and preprodynorphin messenger RNA expression in rat forebrain , 1995, Neuroscience.

[4]  Y. Benjamini,et al.  Controlling the false discovery rate: a practical and powerful approach to multiple testing , 1995 .

[5]  K. Neve,et al.  Dopamine Receptors , 2008 .

[6]  J. Price,et al.  Glial reduction in the subgenual prefrontal cortex in mood disorders. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[7]  B. Roth,et al.  Morphometric evidence for neuronal and glial prefrontal cell pathology in major depression∗ ∗ See accompanying Editorial, in this issue. , 1999, Biological Psychiatry.

[8]  E. Miller,et al.  THE PREFRONTAL CORTEX AND COGNITIVE CONTROL , 2000 .

[9]  E. Miller,et al.  The prefontral cortex and cognitive control , 2000, Nature Reviews Neuroscience.

[10]  J. Cadet,et al.  Temporal profiling of methamphetamine‐induced changes in gene expression in the mouse brain: Evidence from cDNA array , 2001, Synapse.

[11]  R. Kerwin,et al.  Reduced glial cell density and neuronal size in the anterior cingulate cortex in major depressive disorder. , 2001, Archives of general psychiatry.

[12]  J. Zwiller,et al.  Induction of the immediate early genes egr-1 and c-fos by methamphetamine in mouse brain , 2001, Brain Research.

[13]  Xudong Wei,et al.  MEF2C regulates c-Jun but not TNF-alpha gene expression in stimulated mast cells. , 2003, European journal of immunology.

[14]  Xudong Wei,et al.  MEF2C regulates c‐Jun but not TNF‐α gene expression in stimulated mast cells , 2003 .

[15]  R. Fisher,et al.  RGS6 Interacts with DMAP1 and DNMT1 and Inhibits DMAP1 Transcriptional Repressor Activity* , 2004, Journal of Biological Chemistry.

[16]  Jean YH Yang,et al.  Bioconductor: open software development for computational biology and bioinformatics , 2004, Genome Biology.

[17]  Bart De Moor,et al.  BioMart and Bioconductor: a powerful link between biological databases and microarray data analysis , 2005, Bioinform..

[18]  N. Jareborg,et al.  Human QKI, a potential regulator of mRNA expression of human oligodendrocyte-related genes involved in schizophrenia. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[19]  Jonathan Pevsner,et al.  DNA methylation signatures within the human brain. , 2007, American journal of human genetics.

[20]  A. Feinberg Phenotypic plasticity and the epigenetics of human disease , 2007, Nature.

[21]  John D. Storey,et al.  Capturing Heterogeneity in Gene Expression Studies by Surrogate Variable Analysis , 2007, PLoS genetics.

[22]  O. Britanova,et al.  Satb2 Is a Postmitotic Determinant for Upper-Layer Neuron Specification in the Neocortex , 2008, Neuron.

[23]  Clifford A. Meyer,et al.  Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.

[24]  P. Greengard,et al.  Cocaine Regulates MEF2 to Control Synaptic and Behavioral Plasticity , 2008, Neuron.

[25]  S. Mcconnell,et al.  Satb2 Regulates Callosal Projection Neuron Identity in the Developing Cerebral Cortex , 2008, Neuron.

[26]  Mi-Sung Kim,et al.  MEF2C, a transcription factor that facilitates learning and memory by negative regulation of synapse numbers and function , 2008, Proceedings of the National Academy of Sciences.

[27]  Robert Gentleman,et al.  rtracklayer: an R package for interfacing with genome browsers , 2009, Bioinform..

[28]  Hadley Wickham,et al.  ggplot2 - Elegant Graphics for Data Analysis (2nd Edition) , 2017 .

[29]  M. Robinson,et al.  A scaling normalization method for differential expression analysis of RNA-seq data , 2010, Genome Biology.

[30]  Martin J Aryee,et al.  Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts , 2009, Nature Genetics.

[31]  E. Birney,et al.  Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt , 2009, Nature Protocols.

[32]  G. Turecki,et al.  Through the looking glass: examining neuroanatomical evidence for cellular alterations in major depression. , 2009, Journal of psychiatric research.

[33]  A. Feinberg,et al.  Genome-wide methylation analysis of human colon cancer reveals similar hypo- and hypermethylation at conserved tissue-specific CpG island shores , 2008, Nature Genetics.

[34]  Danielle L. Graham,et al.  Striatal regulation of ΔFosB, FosB, and cFos during cocaine self‐administration and withdrawal , 2010, Journal of neurochemistry.

[35]  Christoph Plass,et al.  Tissue specific DNA methylation of CpG islands in normal human adult somatic tissues distinguishes neural from non-neural tissues , 2010, Epigenetics.

[36]  Cory Y. McLean,et al.  GREAT improves functional interpretation of cis-regulatory regions , 2010, Nature Biotechnology.

[37]  E. Klein,et al.  DNA methylation in vulnerability to post-traumatic stress in rats: evidence for the role of the post-synaptic density protein Dlgap2. , 2010, The international journal of neuropsychopharmacology.

[38]  Ayellet V. Segrè,et al.  Hundreds of variants clustered in genomic loci and biological pathways affect human height , 2010, Nature.

[39]  Tanya M. Teslovich,et al.  Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index , 2010 .

[40]  Tanya M. Teslovich,et al.  Biological, Clinical, and Population Relevance of 95 Loci for Blood Lipids , 2010, Nature.

[41]  Ming D. Li,et al.  Genome-wide meta-analyses identify multiple loci associated with smoking behavior , 2010, Nature Genetics.

[42]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[43]  R. Chatterjee,et al.  CpG methylation of half-CRE sequences creates C/EBPα binding sites that activate some tissue-specific genes , 2010, Proceedings of the National Academy of Sciences.

[44]  D. Geschwind,et al.  Neurons show distinctive DNA methylation profile and higher interindividual variations compared with non-neurons. , 2011, Genome research.

[45]  Madeleine P. Ball,et al.  Neuronal activity modifies DNA methylation landscape in the adult brain , 2011, Nature Neuroscience.

[46]  Manuel A. R. Ferreira,et al.  Large-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4 , 2011, Nature Genetics.

[47]  B. Korf,et al.  Clinically relevant single gene or intragenic deletions encompassing critical neurodevelopmental genes in patients with developmental delay, mental retardation, and/or autism spectrum disorders , 2011, American journal of medical genetics. Part A.

[48]  Felix Krueger,et al.  Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications , 2011, Bioinform..

[49]  A. Feinberg,et al.  Increased methylation variation in epigenetic domains across cancer types , 2011, Nature Genetics.

[50]  Hiroyuki Okuno,et al.  Regulation and function of immediate-early genes in the brain: Beyond neuronal activity markers , 2011, Neuroscience Research.

[51]  Disorder Working Group Large-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4 , 2012, Nature Genetics.

[52]  Bronwen L. Aken,et al.  GENCODE: The reference human genome annotation for The ENCODE Project , 2012, Genome research.

[53]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[54]  R. Wise,et al.  Synaptic and Behavioral Profile of Multiple Glutamatergic Inputs to the Nucleus Accumbens , 2012, Neuron.

[55]  B. Langmead,et al.  BSmooth: from whole genome bisulfite sequencing reads to differentially methylated regions , 2012, Genome Biology.

[56]  Claude Bouchard,et al.  A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance , 2012, Nature Genetics.

[57]  R. Dobson,et al.  Functional annotation of the human brain methylome identifies tissue-specific epigenetic variation across brain and blood , 2012, Genome Biology.

[58]  Tanya M. Teslovich,et al.  Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes , 2012, Nature Genetics.

[59]  J. Pasterkamp,et al.  Getting neural circuits into shape with semaphorins , 2012, Nature Reviews Neuroscience.

[60]  K. Becker,et al.  Methamphetamine Causes Differential Alterations in Gene Expression and Patterns of Histone Acetylation/Hypoacetylation in the Rat Nucleus Accumbens , 2012, PloS one.

[61]  Jianxin Shi,et al.  Genetic relationship between five psychiatric disorders estimated from genome-wide SNPs , 2013, Nature Genetics.

[62]  Martin J. Aryee,et al.  Epigenome-wide association data implicate DNA methylation as an intermediary of genetic risk in Rheumatoid Arthritis , 2013, Nature Biotechnology.

[63]  Martin J. Aryee,et al.  A cell epigenotype specific model for the correction of brain cellular heterogeneity bias and its application to age, brain region and major depression , 2013, Epigenetics.

[64]  Jonathan P. Beauchamp,et al.  GWAS of 126,559 Individuals Identifies Genetic Variants Associated with Educational Attainment , 2013, Science.

[65]  R. Irizarry,et al.  Accounting for cellular heterogeneity is critical in epigenome-wide association studies , 2014, Genome Biology.

[66]  Matthew D. Schultz,et al.  Global Epigenomic Reconfiguration During Mammalian Brain Development , 2013, Science.

[67]  Nick C Fox,et al.  Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease , 2013, Nature Genetics.

[68]  J. Qian,et al.  DNA methylation presents distinct binding sites for human transcription factors , 2013, eLife.

[69]  A. Feinberg,et al.  Measuring cell-type specific differential methylation in human brain tissue , 2013, Genome Biology.

[70]  Marni J. Falk,et al.  MEF2C Haploinsufficiency features consistent hyperkinesis, variable epilepsy, and has a role in dorsal and ventral neuronal developmental pathways , 2013, neurogenetics.

[71]  Charity W. Law,et al.  voom: precision weights unlock linear model analysis tools for RNA-seq read counts , 2014, Genome Biology.

[72]  David Haussler,et al.  The UCSC genome browser and associated tools , 2012, Briefings Bioinform..

[73]  Robert Gentleman,et al.  Software for Computing and Annotating Genomic Ranges , 2013, PLoS Comput. Biol..

[74]  Howard Y. Chang,et al.  Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position , 2013, Nature Methods.

[75]  D. Drapier,et al.  The nucleus accumbens: a target for deep brain stimulation in resistant major depressive disorder , 2013, Journal of molecular psychiatry.

[76]  Minghua Wu,et al.  Function and mechanism of tumor suppressor gene LRRC4/NGL-2 , 2014, Molecular Cancer.

[77]  C. Spencer,et al.  Biological Insights From 108 Schizophrenia-Associated Genetic Loci , 2014, Nature.

[78]  T. Schlaepfer,et al.  Aberrant NMDA receptor DNA methylation detected by epigenome-wide analysis of hippocampus and prefrontal cortex in major depression , 2015, European Archives of Psychiatry and Clinical Neuroscience.

[79]  S. Orkin,et al.  Analysis of chromatin-state plasticity identifies cell-type–specific regulators of H3K27me3 patterns , 2014, Proceedings of the National Academy of Sciences.

[80]  R Core Team,et al.  R: A language and environment for statistical computing. , 2014 .

[81]  A. Feinberg,et al.  Large-scale hypomethylated blocks associated with Epstein-Barr virus–induced B-cell immortalization , 2014, Genome research.

[82]  M. Creyghton,et al.  Large-scale identification of coregulated enhancer networks in the adult human brain. , 2014, Cell reports.

[83]  T. Meehan,et al.  An atlas of active enhancers across human cell types and tissues , 2014, Nature.

[84]  Andrew D. Johnson,et al.  Parent-of-origin specific allelic associations among 106 genomic loci for age at menarche , 2014, Nature.

[85]  Jun S. Liu,et al.  Genetics of rheumatoid arthritis contributes to biology and drug discovery , 2013 .

[86]  Gustavo Turecki,et al.  Methylomic profiling of human brain tissue supports a neurodevelopmental origin for schizophrenia , 2014, Genome Biology.

[87]  W. Fu,et al.  Autism-associated gene Dlgap2 mutant mice demonstrate exacerbated aggressive behaviors and orbitofrontal cortex deficits , 2014, Molecular Autism.

[88]  Ian J. Deary,et al.  Common genetic variants associated with cognitive performance identified using the proxy-phenotype method , 2014, Proceedings of the National Academy of Sciences.

[89]  E. Koonin,et al.  Differences in DNA methylation between human neuronal and glial cells are concentrated in enhancers and non-CpG sites , 2013, Nucleic acids research.

[90]  Gokhan Guner,et al.  Genome-wide target analysis of NEUROD2 provides new insights into regulation of cortical projection neuron migration and differentiation , 2015, BMC Genomics.

[91]  Dmitri D. Pervouchine,et al.  The human transcriptome across tissues and individuals , 2015, Science.

[92]  C. Allerston,et al.  Structures of the Ets Protein DNA-binding Domains of Transcription Factors Etv1, Etv4, Etv5, and Fev , 2015, The Journal of Biological Chemistry.

[93]  I. Mavridis [The role of the nucleus accumbens in psychiatric disorders]. , 2015, Psychiatrike = Psychiatriki.

[94]  Robert Andrews,et al.  Inter-individual variability contrasts with regional homogeneity in the human brain DNA methylome , 2015, Nucleic acids research.

[95]  O. Lopez,et al.  Genetic variation in imprinted genes is associated with risk of late-onset Alzheimer's disease. , 2015, Journal of Alzheimer's disease : JAD.

[96]  Matthew E. Ritchie,et al.  limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.

[97]  Terrence J. Sejnowski,et al.  Epigenomic Signatures of Neuronal Diversity in the Mammalian Brain , 2015, Neuron.

[98]  Andrew P. Feinberg,et al.  An LSC epigenetic signature is largely mutation independent and implicates the HOXA cluster in AML pathogenesis , 2015, Nature Communications.

[99]  Raphael Gottardo,et al.  Orchestrating high-throughput genomic analysis with Bioconductor , 2015, Nature Methods.

[100]  Yakir A Reshef,et al.  Partitioning heritability by functional annotation using genome-wide association summary statistics , 2015, Nature Genetics.

[101]  Morris Swertz,et al.  Genome-wide patterns and properties of de novo mutations in humans , 2015, Nature Genetics.

[102]  M. Daly,et al.  LD Score regression distinguishes confounding from polygenicity in genome-wide association studies , 2014, Nature Genetics.

[103]  Howard Y. Chang,et al.  ATAC‐seq: A Method for Assaying Chromatin Accessibility Genome‐Wide , 2015, Current protocols in molecular biology.

[104]  M. Robinson,et al.  Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences , 2015, F1000Research.

[105]  M. Kaplitt,et al.  The Nucleus Accumbens: A Comprehensive Review , 2015, Stereotactic and Functional Neurosurgery.

[106]  Chia-Hsiang Chen,et al.  Resequencing of early growth response 2 (EGR2) gene revealed a recurrent patient-specific mutation in schizophrenia , 2015, Psychiatry Research.

[107]  Michael Q. Zhang,et al.  Integrative analysis of 111 reference human epigenomes , 2015, Nature.

[108]  C. Coarfa,et al.  CpG Methylation Differences Between Neurons and Glia are Highly Conserved from Mouse to Human , 2016, Human molecular genetics.

[109]  Samuel S. Gross,et al.  Genome-wide characteristics of de novo mutations in autism , 2016, npj Genomic Medicine.

[110]  O. Lazarov,et al.  Alzheimer's Disease and Hippocampal Adult Neurogenesis; Exploring Shared Mechanisms , 2016, Front. Neurosci..

[111]  F. Piras,et al.  Left nucleus accumbens atrophy in deficit schizophrenia: A preliminary study , 2016, Psychiatry Research: Neuroimaging.

[112]  F. Klamt,et al.  Differential expression of transcriptional regulatory units in the prefrontal cortex of patients with bipolar disorder: potential role of early growth response gene 3 , 2016, Translational psychiatry.

[113]  Jiang Qian,et al.  Transcription factors as readers and effectors of DNA methylation , 2016, Nature Reviews Genetics.

[114]  E. Maguire,et al.  Anterior hippocampus: the anatomy of perception, imagination and episodic memory , 2016, Nature Reviews Neuroscience.

[115]  S. Ropero,et al.  Epigenetics in Schizophrenia: A Pilot Study of Global DNA Methylation in Different Brain Regions Associated with Higher Cognitive Functions , 2016, Front. Psychol..

[116]  Lei Wang,et al.  Subcortical neuromorphometry in schizophrenia spectrum and bipolar disorders , 2016, NeuroImage: Clinical.

[117]  I. Csabai,et al.  Aberrant DNA methylation of WNT pathway genes in the development and progression of CIMP-negative colorectal cancer , 2016, Epigenetics.

[118]  Brian T. Lee,et al.  The UCSC Genome Browser database: 2015 update , 2014, Nucleic Acids Res..

[119]  David J. Arenillas,et al.  JASPAR 2016: a major expansion and update of the open-access database of transcription factor binding profiles , 2015, Nucleic Acids Res..

[120]  Andrew D. Rouillard,et al.  Enrichr: a comprehensive gene set enrichment analysis web server 2016 update , 2016, Nucleic Acids Res..

[121]  P. Kalivas,et al.  The Nucleus Accumbens: Mechanisms of Addiction across Drug Classes Reflect the Importance of Glutamate Homeostasis , 2016, Pharmacological Reviews.

[122]  Kyle J Gerber,et al.  Roles for Regulator of G Protein Signaling Proteins in Synaptic Signaling and Plasticity , 2016, Molecular Pharmacology.

[123]  Jonathan P. Beauchamp,et al.  Genetic variants associated with subjective well-being, depressive symptoms and neuroticism identified through genome-wide analyses , 2016, Nature Genetics.

[124]  Daniel R Weinberger,et al.  Mapping DNA methylation across development, genotype, and schizophrenia in the human frontal cortex , 2015, Nature Neuroscience.

[125]  Geet Duggal,et al.  Salmon provides accurate, fast, and bias-aware transcript expression estimates using dual-phase inference , 2015, bioRxiv.

[126]  J. Mill,et al.  Schizophrenia-associated methylomic variation: molecular signatures of disease and polygenic risk burden across multiple brain regions , 2016, Human molecular genetics.

[127]  G. Ming,et al.  Neuronal activity modifies the chromatin accessibility landscape in the adult brain , 2017, Nature Neuroscience.