Functional annotation of the human brain methylome identifies tissue-specific epigenetic variation across brain and blood

Matthew N Davies, Manuela Volta, Ruth Pidsley, Katie Lunnon, Abhishek Dixit, Simon Lovestone, Cristian Coarfa, R Alan Harris, Aleksandar Milosavljevic, Claire Troakes, Safa Al-Sarraj, Richard Dobson, Leonard C Schalkwyk, Jonathan Mill, Matthew N Davies, Manuela Volta, Ruth Pidsley, Katie Lunnon, Abhishek Dixit, Simon Lovestone, Cristian Coarfa, R Alan Harris, Aleksandar Milosavljevic, Claire Troakes, Safa Al-Sarraj, Richard Dobson, Leonard C Schalkwyk, Jonathan Mill

Abstract

Background: Dynamic changes to the epigenome play a critical role in establishing and maintaining cellular phenotype during differentiation, but little is known about the normal methylomic differences that occur between functionally distinct areas of the brain. We characterized intra- and inter-individual methylomic variation across whole blood and multiple regions of the brain from multiple donors.

Results: Distinct tissue-specific patterns of DNA methylation were identified, with a highly significant over-representation of tissue-specific differentially methylated regions (TS-DMRs) observed at intragenic CpG islands and low CG density promoters. A large proportion of TS-DMRs were located near genes that are differentially expressed across brain regions. TS-DMRs were significantly enriched near genes involved in functional pathways related to neurodevelopment and neuronal differentiation, including BDNF, BMP4, CACNA1A, CACA1AF, EOMES, NGFR, NUMBL, PCDH9, SLIT1, SLITRK1 and SHANK3. Although between-tissue variation in DNA methylation was found to greatly exceed between-individual differences within any one tissue, we found that some inter-individual variation was reflected across brain and blood, indicating that peripheral tissues may have some utility in epidemiological studies of complex neurobiological phenotypes.

Conclusions: This study reinforces the importance of DNA methylation in regulating cellular phenotype across tissues, and highlights genomic patterns of epigenetic variation across functionally distinct regions of the brain, providing a resource for the epigenetics and neuroscience research communities.

Figures

Figure 1
Figure 1
Methylomic profiling across multiple brain areas and blood from a cohort of individuals highlights clear tissue-specific differences in DNA methylation. (a, b) DNA methylation was calculated from ultra-deep MeDIP-seq data using 500 bp bins across the genome, and the relationship between tissues determined by Pearson correlation (a) and unsupervised hierarchical clustering (b). BA, Brodmann area; Ent Ctx, entorhinal cortex; STG, superior temporal gyrus; Vis Ctx, visual cortex.
Figure 2
Figure 2
Although DNA methylation at CGIs is relatively conserved across tissues, intragenic CGIs are dramatically over-represented and promoter CGIs under-represented in the most tissue-variable CGIs. (a) Average DNA methylation values calculated by MEDIPS from MeDIP-seq data for all annotated gene features: CGIs (yellow), CGI shores (blue), gene promoters (red) and CDSs (green). DNA methylation is lower in promoter CGIs compared to intragenic, 3' UTR and intergenic CGIs. CGI shores are characterized by higher DNA methylation than CGIs, with less location-dependent variation. Promoter DNA methylation shows a strong inverse correlation with GC density, with LCPs showing a higher average level of DNA methylation than CDSs. Error bars represent standard error of the mean. (b) Although TS-DMRs are distributed across all feature types, there are marked differences in the between-tissue correlation of DNA methylation across each of the broad feature categories we examined, with CGIs being more correlated across cortex, cerebellum and blood than CGI shores or CDSs. (c, d) There is a highly significant enrichment of intragenic CGIs (P = 2 × 10-102) in analyses of CGI DMRs differentiating blood, cortex and cerebellum (c), and an even more dramatic enrichment (P = 1 × 10-246) in comparisons between cortex and cerebellum (d). EXP, expected; OBS, observed.
Figure 3
Figure 3
Verification and replication of MeDIP-seq data for three top-ranked CGI DMRs. (a, b) Tissue-specific DNA methylation across an intragenic CGI in the JMJD2B/KDM4B gene. (a) MeDIP-seq analysis shows this region is hypermethylated in blood DNA compared to cortex and cerebellum (the red bar depicts the region subsequently analyzed by bisulfite pyrosequencing). (b) Pyrosequencing data for this region in an extended sample set confirm significant tissue-specific methylation patterns (P = 2 × 10-8). (c, d) Tissue-specific DNA methylation across a CGI in the promoter of the EOMES gene. (c) MeDIP-seq analysis shows this region is hypermethylated in cerebellum DNA compared to cortex and blood. (d) Pyrosequencing data for this region in an extended sample set confirm significant tissue-specific methylation patterns (P = 2 × 10-5). (e, f) Tissue-specific DNA methylation across an intragenic CGI in the BDNF gene. (e) MeDIP-seq analysis shows this region is hypermethylated in blood DNA compared to cortex and cerebellum from the same individuals. (f) Pyrosequencing data for this region in an extended sample set confirm significant tissue-specific methylation patterns (P = 4 × 10-9). Error bars represent standard error of the mean.
Figure 4
Figure 4
Weighted gene co-methylation network analysis of DNA methylation at intragenic CGIs. (a) Dendrograms produced by average linkage hierarchical clustering of intragenic CGIs on the basis of topological overlap. Modules of co-methylated loci were assigned colors as indicated by the horizontal bar beneath each dendrogram. The 'blue' module was strongly negatively co-methylated (r2 = -0.98, P = 4 × 10-5) in cortex. (b) IPA on the genes associated with the blue module highlighted a network involved in nervous system development and function.
Figure 5
Figure 5
DNA methylation across LCPs is strongly associated with tissue type. (a) MEDIPS scores across both HCPs and LCPs can be used to accurately cluster samples by tissue type, but the strength of clustering, indicated by Pearson dissimilarity on the y-axis, is much higher in LCPs. (b, c) This pattern is reflected in three-factor PCA plots (b) and correlation analyses (c), with LCPs demonstrating stronger tissue-specific patterns of DNA methylation than HCPs.
Figure 6
Figure 6
Between-individual variation in DNA methylation is often correlated between blood and brain. (a) Between-individual variation in DNA methylation is highest in blood and lowest in the cortex. All tissues show a similarly significant (ANOVA P < 0.001) distribution of variability across features, with the greatest between-individual variation occurring in non-promoter CGIs. Scores represent the mean difference in normalized MeDIP-seq read density between individual 1 and 2 for each of the feature categories. Error bars represent standard error of the mean. Features 1 to 4 = promoter, intragenic, 3', and intergenic CGIs; features 4 to 8 = promoter, intragenic, 3', and intergenic CGI shores; feature 9 = CDS; features 10 to 12 = HCPs, ICPs, and LCPs. (b) Between-individual differences in DNA methylation observed in blood are significantly (P < 0.001) correlated with differences observed in the cerebellum (correlation = 0.76) and cortex (correlation = 0.66) from the same individuals. Scores represent the mean difference in normalized MeDIP-seq read density between individual 1 and 2 for each of the quantified features.

References

    1. Suzuki MM, Bird A. DNA methylation landscapes: provocative insights from epigenomics. Nat Rev Genet. 2008;9:465–476.
    1. Ma DK, Marchetto MC, Guo JU, Ming GL, Gage FH, Song H. Epigenetic choreographers of neurogenesis in the adult mammalian brain. Nat Neurosci. 2010;13:1338–1344. doi: 10.1038/nn.2672.
    1. Guo JU, Ma DK, Mo H, Ball MP, Jang MH, Bonaguidi MA, Balazer JA, Eaves HL, Xie B, Ford E, Zhang K, Ming GL, Gao Y, Song H. Neuronal activity modifies the DNA methylation landscape in the adult brain. Nat Neurosci. 2011;14:1345–1351. doi: 10.1038/nn.2900.
    1. Lubin FD, Roth TL, Sweatt JD. Epigenetic regulation of BDNF gene transcription in the consolidation of fear memory. J Neurosci. 2008;28:10576–10586. doi: 10.1523/JNEUROSCI.1786-08.2008.
    1. Renthal W, Nestler EJ. Histone acetylation in drug addiction. Semin Cell Dev Biol. 2009;20:387–394. doi: 10.1016/j.semcdb.2009.01.005.
    1. Migliore L, Coppede F. Genetics, environmental factors and the emerging role of epigenetics in neurodegenerative diseases. Mutat Res. 2009;667:82–97. doi: 10.1016/j.mrfmmm.2008.10.011.
    1. Nakahata Y, Grimaldi B, Sahar S, Hirayama J, Sassone-Corsi P. Signaling to the circadian clock: plasticity by chromatin remodeling. Curr Opin Cell Biol. 2007;19:230–237. doi: 10.1016/j.ceb.2007.02.016.
    1. Samaco RC, Neul JL. Complexities of Rett syndrome and MeCP2. J Neurosci. 2011;31:7951–7959. doi: 10.1523/JNEUROSCI.0169-11.2011.
    1. Mill J, Tang T, Kaminsky Z, Khare T, Yazdanpanah S, Bouchard L, Jia P, Assadzadeh A, Flanagan J, Schumacher A, Wang SC, Petronis A. Epigenomic profiling reveals DNA-methylation changes associated with major psychosis. Am J Hum Genet. 2008;82:696–711. doi: 10.1016/j.ajhg.2008.01.008.
    1. Roth RB, Hevezi P, Lee J, Willhite D, Lechner SM, Foster AC, Zlotnik A. Gene expression analyses reveal molecular relationships among 20 regions of the human CNS. Neurogenetics. 2006;7:67–80. doi: 10.1007/s10048-006-0032-6.
    1. Khaitovich P, Muetzel B, She X, Lachmann M, Hellmann I, Dietzsch J, Steigele S, Do HH, Weiss G, Enard W, Heissig F, Arendt T, Nieselt-Struwe K, Eichler EE, Paabo S. Regional patterns of gene expression in human and chimpanzee brains. Genome Res. 2004;14:1462–1473. doi: 10.1101/gr.2538704.
    1. Johnson MB, Kawasawa YI, Mason CE, Krsnik Z, Coppola G, Bogdanovic D, Geschwind DH, Mane SM, State MW, Sestan N. Functional and evolutionary insights into human brain development through global transcriptome analysis. Neuron. 2009;62:494–509. doi: 10.1016/j.neuron.2009.03.027.
    1. Ladd-Acosta C, Pevsner J, Sabunciyan S, Yolken RH, Webster MJ, Dinkins T, Callinan PA, Fan JB, Potash JB, Feinberg AP. DNA methylation signatures within the human brain. Am J Hum Genet. 2007;81:1304–1315. doi: 10.1086/524110.
    1. Chavez L, Jozefczuk J, Grimm C, Dietrich J, Timmermann B, Lehrach H, Herwig R, Adjaye J. Computational analysis of genome-wide DNA methylation during the differentiation of human embryonic stem cells along the endodermal lineage. Genome Res. 2010;20:1441–1450. doi: 10.1101/gr.110114.110.
    1. KCL Psychiatric Epigenetics Data.
    1. MeDIP-seq data page in Human Epigenome Atlas.
    1. Human Epigenome Atlas.
    1. Bernstein BE, Stamatoyannopoulos JA, Costello JF, Ren B, Milosavljevic A, Meissner A, Kellis M, Marra MA, Beaudet AL, Ecker JR, Farnham PJ, Hirst M, Lander ES, Mikkelsen TS, Thomson JA. The NIH Roadmap Epigenomics Mapping Consortium. Nat Biotechnol. 2010;28:1045–1048. doi: 10.1038/nbt1010-1045.
    1. Maunakea AK, Nagarajan RP, Bilenky M, Ballinger TJ, D'Souza C, Fouse SD, Johnson BE, Hong C, Nielsen C, Zhao Y, Turecki G, Delaney A, Varhol R, Thiessen N, Shchors K, Heine VM, Rowitch DH, Xing X, Fiore C, Schillebeeckx M, Jones SJ, Haussler D, Marra MA, Hirst M, Wang T, Costello JF. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature. 2010;466:253–257. doi: 10.1038/nature09165.
    1. Weber M, Hellmann I, Stadler MB, Ramos L, Paabo S, Rebhan M, Schubeler D. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat Genet. 2007;39:457–466. doi: 10.1038/ng1990.
    1. Doi A, Park IH, Wen B, Murakami P, Aryee MJ, Irizarry R, Herb B, Ladd-Acosta C, Rho J, Loewer S, Miller J, Schlaeger T, Daley GQ, Feinberg AP. Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nat Genet. 2009;41:1350–1353. doi: 10.1038/ng.471.
    1. Sato K, Yabe I, Fukuda Y, Soma H, Nakahara Y, Tsuji S, Sasaki H. Mapping of autosomal dominant cerebellar ataxia without the pathogenic PPP2R2B mutation to the locus for spinocerebellar ataxia 12. Arch Neurol. 2010;67:1257–1262. doi: 10.1001/archneurol.2010.231.
    1. Couve A, Restituito S, Brandon JM, Charles KJ, Bawagan H, Freeman KB, Pangalos MN, Calver AR, Moss SJ. Marlin-1, a novel RNA-binding protein associates with GABA receptors. J Biol Chem. 2004;279:13934–13943. doi: 10.1074/jbc.M311737200.
    1. Baala L, Briault S, Etchevers HC, Laumonnier F, Natiq A, Amiel J, Boddaert N, Picard C, Sbiti A, Asermouh A, Attie-Bitach T, Encha-Razavi F, Munnich A, Sefiani A, Lyonnet S. Homozygous silencing of T-box transcription factor EOMES leads to microcephaly with polymicrogyria and corpus callosum agenesis. Nat Genet. 2007;39:454–456. doi: 10.1038/ng1993.
    1. Werner H, Dimou L, Klugmann M, Pfeiffer S, Nave KA. Multiple splice isoforms of proteolipid M6B in neurons and oligodendrocytes. Mol Cell Neurosci. 2001;18:593–605. doi: 10.1006/mcne.2001.1044.
    1. Makoff A, Lelchuk R, Oxer M, Harrington K, Emson P. Molecular characterization and localization of human metabotropic glutamate receptor type 4. Brain Res Mol Brain Res. 1996;37:239–248.
    1. Ramakers GJ, Avci B, van Hulten P, van Ooyen A, van Pelt J, Pool CW, Lequin MB. The role of calcium signaling in early axonal and dendritic morphogenesis of rat cerebral cortex neurons under non-stimulated growth conditions. Brain Res Dev Brain Res. 2001;126:163–172.
    1. Toba S, Tenno M, Konishi M, Mikami T, Itoh N, Kurosaka A. Brain-specific expression of a novel human UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferase (GalNAc-T9). Biochim Biophys Acta. 2000;1493:264–268.
    1. Gibney GT, Zhang JH, Douglas RM, Haddad GG, Xia Y. Na(+)/Ca(2+) exchanger expression in the developing rat cortex. Neuroscience. 2002;112:65–73. doi: 10.1016/S0306-4522(02)00059-3.
    1. Zhong W, Jiang MM, Weinmaster G, Jan LY, Jan YN. Differential expression of mammalian Numb, Numblike and Notch1 suggests distinct roles during mouse cortical neurogenesis. Development. 1997;124:1887–1897.
    1. Hayes DM, Braud S, Hurtado DE, McCallum J, Standley S, Isaac JT, Roche KW. Trafficking and surface expression of the glutamate receptor subunit, KA2. Biochem Biophys Res Commun. 2003;310:8–13. doi: 10.1016/j.bbrc.2003.08.115.
    1. Sun Y, Hu J, Zhou L, Pollard SM, Smith A. Interplay between FGF2 and BMP controls the self-renewal, dormancy and differentiation of rat neural stem cells. J Cell Sci. 2011;124:1867–1877. doi: 10.1242/jcs.085506.
    1. Boulle F, van den Hove DL, Jakob SB, Rutten BP, Hamon M, van Os J, Lesch KP, Lanfumey L, Steinbusch HW, Kenis G. Epigenetic regulation of the BDNF gene: implications for psychiatric disorders. Mol Psychiatry. 2011;17:584–596.
    1. Kajiwara Y, Buxbaum JD, Grice DE. SLITRK1 binds 14-3-3 and regulates neurite outgrowth in a phosphorylation-dependent manner. Biol Psychiatry. 2009;66:918–925. doi: 10.1016/j.biopsych.2009.05.033.
    1. Zhang B, Horvath S. A general framework for weighted gene co-expression network analysis. Stat Appl Genet Mol Biol. 2005;4:Article17.
    1. GeneMANIA.
    1. Deaton AM, Webb S, Kerr AR, Illingworth RS, Guy J, Andrews R, Bird A. Cell type-specific DNA methylation at intragenic CpG islands in the immune system. Genome Res. 2011;21:1074–1086. doi: 10.1101/gr.118703.110.
    1. Kim SY, Mo JW, Han S, Choi SY, Han SB, Moon BH, Rhyu IJ, Sun W, Kim H. The expression of non-clustered protocadherins in adult rat hippocampal formation and the connecting brain regions. Neuroscience. 2010;170:189–199. doi: 10.1016/j.neuroscience.2010.05.027.
    1. Cosgaya JM, Chan JR, Shooter EM. The neurotrophin receptor p75NTR as a positive modulator of myelination. Science. 2002;298:1245–1248. doi: 10.1126/science.1076595.
    1. Bedogni F, Hodge RD, Nelson BR, Frederick EA, Shiba N, Daza RA, Hevner RF. Autism susceptibility candidate 2 (Auts2) encodes a nuclear protein expressed in developing brain regions implicated in autism neuropathology. Gene Expr Patterns. 2010;10:9–15. doi: 10.1016/j.gep.2009.11.005.
    1. Herbert MR. SHANK3, the synapse, and autism. N Engl J Med. pp. 173–175.
    1. Whitford KL, Marillat V, Stein E, Goodman CS, Tessier-Lavigne M, Chedotal A, Ghosh A. Regulation of cortical dendrite development by Slit-Robo interactions. Neuron. 2002;33:47–61. doi: 10.1016/S0896-6273(01)00566-9.
    1. Borgel J, Guibert S, Li Y, Chiba H, Schubeler D, Sasaki H, Forne T, Weber M. Targets and dynamics of promoter DNA methylation during early mouse development. Nat Genet. pp. 1093–1100.
    1. Petronis A. Epigenetics as a unifying principle in the aetiology of complex traits and diseases. Nature. 2010;465:721–727. doi: 10.1038/nature09230.
    1. Davies MN, Lawn S, Whatley S, Fernandes C, Williams RW, Schalkwyk LC. To what extent is blood a reasonable surrogate for brain in gene expression studies: estimation from mouse hippocampus and spleen. Front Neurosci. 2009;3:54.
    1. Rakyan VK, Down TA, Balding DJ, Beck S. Epigenome-wide association studies for common human diseases. Nat Rev Genet. 2011;12:529–541. doi: 10.1038/nrg3000.
    1. Schalkwyk LC, Meaburn EL, Smith R, Dempster EL, Jeffries AR, Davies MN, Plomin R, Mill J. Allelic skewing of DNA methylation is widespread across the genome. Am J Hum Genet. 2010;86:196–212. doi: 10.1016/j.ajhg.2010.01.014.
    1. MAQ: Mapping and Assembly with Qualities.
    1. FastQC.
    1. Horvath S, Dong J. Geometric interpretation of gene coexpression network analysis. PLoS Comput Biol. 2008;4:e1000117. doi: 10.1371/journal.pcbi.1000117.
    1. Horvath S, Zhang B, Carlson M, Lu KV, Zhu S, Felciano RM, Laurance MF, Zhao W, Qi S, Chen Z, Lee Y, Scheck AC, Liau LM, Wu H, Geschwind DH, Febbo PG, Kornblum HI, Cloughesy TF, Nelson SF, Mischel PS. Analysis of oncogenic signaling networks in glioblastoma identifies ASPM as a molecular target. Proc Natl Acad Sci USA. 2006;103:17402–17407. doi: 10.1073/pnas.0608396103.
    1. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559. doi: 10.1186/1471-2105-9-559.
    1. PennCNV.

Source: PubMed

3
Abonneren