Integrative functional genomic analysis of human brain development and neuropsychiatric risks

INTRODUCTION The brain is responsible for cognition, behavior, and much of what makes us uniquely human. The development of the brain is a highly complex process, and this process is reliant on precise regulation of molecular and cellular events grounded in the spatiotemporal regulation of the transcriptome. Disruption of this regulation can lead to neuropsychiatric disorders. RATIONALE The regulatory, epigenomic, and transcriptomic features of the human brain have not been comprehensively compiled across time, regions, or cell types. Understanding the etiology of neuropsychiatric disorders requires knowledge not just of endpoint differences between healthy and diseased brains but also of the developmental and cellular contexts in which these differences arise. Moreover, an emerging body of research indicates that many aspects of the development and physiology of the human brain are not well recapitulated in model organisms, and therefore it is necessary that neuropsychiatric disorders be understood in the broader context of the developing and adult human brain. RESULTS Here we describe the generation and analysis of a variety of genomic data modalities at the tissue and single-cell levels, including transcriptome, DNA methylation, and histone modifications across multiple brain regions ranging in age from embryonic development through adulthood. We observed a widespread transcriptomic transition beginning during late fetal development and consisting of sharply decreased regional differences. This reduction coincided with increases in the transcriptional signatures of mature neurons and the expression of genes associated with dendrite development, synapse development, and neuronal activity, all of which were temporally synchronous across neocortical areas, as well as myelination and oligodendrocytes, which were asynchronous. Moreover, genes including MEF2C, SATB2, and TCF4, with genetic associations to multiple brain-related traits and disorders, converged in a small number of modules exhibiting spatial or spatiotemporal specificity. CONCLUSION We generated and applied our dataset to document transcriptomic and epigenetic changes across human development and then related those changes to major neuropsychiatric disorders. These data allowed us to identify genes, cell types, gene coexpression modules, and spatiotemporal loci where disease risk might converge, demonstrating the utility of the dataset and providing new insights into human development and disease. Spatiotemporal dynamics of human brain development and neuropsychiatric risks. Human brain development begins during embryonic development and continues through adulthood (top). Integrating data modalities (bottom left) revealed age- and cell type–specific properties and global patterns of transcriptional dynamics, including a late fetal transition (bottom middle). We related the variation in gene expression (brown, high; purple, low) to regulatory elements in the fetal and adult brains, cell type–specific signatures, and genetic loci associated with neuropsychiatric disorders (bottom right; gray circles indicate enrichment for corresponding features among module genes). Relationships depicted in this panel do not correspond to specific observations. CBC, cerebellar cortex; STR, striatum; HIP, hippocampus; MD, mediodorsal nucleus of thalamus; AMY, amygdala. To broaden our understanding of human neurodevelopment, we profiled transcriptomic and epigenomic landscapes across brain regions and/or cell types for the entire span of prenatal and postnatal development. Integrative analysis revealed temporal, regional, sex, and cell type–specific dynamics. We observed a global transcriptomic cup-shaped pattern, characterized by a late fetal transition associated with sharply decreased regional differences and changes in cellular composition and maturation, followed by a reversal in childhood-adolescence, and accompanied by epigenomic reorganizations. Analysis of gene coexpression modules revealed relationships with epigenomic regulation and neurodevelopmental processes. Genes with genetic associations to brain-based traits and neuropsychiatric disorders (including MEF2C, SATB2, SOX5, TCF4, and TSHZ3) converged in a small number of modules and distinct cell types, revealing insights into neurodevelopment and the genomic basis of neuropsychiatric risks.

[1]  Gerome Breen,et al.  Genetic identification of brain cell types underlying schizophrenia , 2017, Nature Genetics.

[2]  Daniel J. Miller,et al.  Spatiotemporal transcriptomic divergence across human and macaque brain development , 2018, Science.

[3]  Prashant S. Emani,et al.  Comprehensive functional genomic resource and integrative model for the human brain , 2018, Science.

[4]  C. d’Enfert Evolving a pathogen to be protective , 2018, Science.

[5]  Alicia R. Martin,et al.  Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder , 2018, Nature Genetics.

[6]  Yun Li,et al.  Schizophrenia and a high-resolution map of the three-dimensional chromatin interactome of adult and fetal cortex , 2018, bioRxiv.

[7]  Tyrone D. Cannon,et al.  Genome-wide association meta-analysis in 269,867 individuals identifies new genetic and functional links to intelligence , 2018, Nature Genetics.

[8]  Mingfeng Li,et al.  Early emergence of cortical interneuron diversity in the mouse embryo , 2018, Science.

[9]  Paul Hoffman,et al.  Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.

[10]  Jie Qiao,et al.  A single-cell RNA-seq survey of the developmental landscape of the human prefrontal cortex , 2018, Nature.

[11]  Jakob Grove,et al.  Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection , 2018, Nature Genetics.

[12]  Warren W. Kretzschmar,et al.  Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression , 2017, Nature Genetics.

[13]  Jason M Keil,et al.  Brain Transcriptome Databases: A User's Guide , 2018, The Journal of Neuroscience.

[14]  D. Geschwind,et al.  The Dynamic Landscape of Open Chromatin during Human Cortical Neurogenesis , 2018, Cell.

[15]  John P. Rice,et al.  Genomic Dissection of Bipolar Disorder and Schizophrenia, Including 28 Subphenotypes , 2017, Cell.

[16]  P. Kharchenko,et al.  Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain , 2017, Nature Biotechnology.

[17]  S. Horvath,et al.  Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap , 2016, Science.

[18]  Alex A. Pollen,et al.  Spatiotemporal gene expression trajectories reveal developmental hierarchies of the human cortex , 2017, Science.

[19]  Ian J. Deary,et al.  Association analysis in over 329,000 individuals identifies 116 independent variants influencing neuroticism , 2017, Nature Genetics.

[20]  Jakob Grove,et al.  Common risk variants identified in autism spectrum disorder , 2017, bioRxiv.

[21]  Christian Gieger,et al.  Impact of common genetic determinants of Hemoglobin A1c on type 2 diabetes risk and diagnosis in ancestrally diverse populations: A transethnic genome-wide meta-analysis , 2017, PLoS Medicine.

[22]  Michael Hawrylycz,et al.  Transcriptomic Perspectives on Neocortical Structure, Development, Evolution, and Disease. , 2017, Annual review of neuroscience.

[23]  Jakob Grove,et al.  Discovery of the first genome-wide significant risk loci for ADHD , 2017, bioRxiv.

[24]  N. Hacohen,et al.  Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors , 2017, Science.

[25]  Joan,et al.  Prevalence and architecture of de novo mutations in developmental disorders , 2017, Nature.

[26]  Deciphering Developmental Disorders Study,et al.  Prevalence and architecture of de novo mutations in developmental disorders , 2017, Nature.

[27]  Can Yang,et al.  Simultaneous dimension reduction and adjustment for confounding variation , 2016, Proceedings of the National Academy of Sciences.

[28]  Daning Lu,et al.  Chromosome conformation elucidates regulatory relationships in developing human brain , 2016, Nature.

[29]  Soher Balkhy,et al.  Mutations in Human Accelerated Regions Disrupt Cognition and Social Behavior , 2016, Cell.

[30]  Genevieve Konopka,et al.  MEF2C regulates cortical inhibitory and excitatory synapses and behaviors relevant to neurodevelopmental disorders , 2016, eLife.

[31]  P. Gubellini,et al.  TSHZ3 deletion causes an autism syndrome and defects in cortical projection neurons , 2016, Nature Genetics.

[32]  D. Ecker,et al.  Tcf4 Regulates Synaptic Plasticity, DNA Methylation, and Memory Function. , 2016, Cell reports.

[33]  M. Ronaghi,et al.  Neuronal subtypes and diversity revealed by single-nucleus RNA sequencing of the human brain , 2016, Science.

[34]  N. Šestan,et al.  The Cellular and Molecular Landscapes of the Developing Human Central Nervous System , 2016, Neuron.

[35]  Tal Pupko,et al.  Genomic analysis of 38 Legionella species identifies large and diverse effector repertoires , 2016, Nature Genetics.

[36]  Paola Arlotta,et al.  Generating neuronal diversity in the mammalian cerebral cortex. , 2015, Annual review of cell and developmental biology.

[37]  Matthias Heinig,et al.  Alternative Splicing Signatures in RNA‐seq Data: Percent Spliced in (PSI) , 2015, Current protocols in human genetics.

[38]  Christopher S. Poultney,et al.  Insights into Autism Spectrum Disorder Genomic Architecture and Biology from 71 Risk Loci , 2015, Neuron.

[39]  Judy H. Cho,et al.  Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations , 2015, Nature Genetics.

[40]  S. Quake,et al.  A survey of human brain transcriptome diversity at the single cell level , 2015, Proceedings of the National Academy of Sciences.

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

[42]  Jun S. Liu,et al.  The Genotype-Tissue Expression (GTEx) pilot analysis: Multitissue gene regulation in humans , 2015, Science.

[43]  E. Schuman,et al.  Direct visualization of newly synthesized target proteins in situ , 2015, Nature Methods.

[44]  Kutay D Atabay,et al.  Single-cell analysis reveals transcriptional heterogeneity of neural progenitors in human cortex , 2015, Nature Neuroscience.

[45]  Janet Kelso,et al.  Human-specific gene ARHGAP11B promotes basal progenitor amplification and neocortex expansion , 2015, Science.

[46]  Ash A. Alizadeh,et al.  Robust enumeration of cell subsets from tissue expression profiles , 2015, Nature Methods.

[47]  Jing Leng,et al.  Evolutionary changes in promoter and enhancer activity during human corticogenesis , 2015, Science.

[48]  Michael C O'Donovan,et al.  Methylomic trajectories across human fetal brain development , 2015, Genome research.

[49]  C. Walsh,et al.  Genetic Changes Shaping the Human Brain , 2015 .

[50]  A. Regev,et al.  Spatial reconstruction of single-cell gene expression , 2015, Nature Biotechnology.

[51]  Nenad Sestan,et al.  From trans to cis: transcriptional regulatory networks in neocortical development. , 2015, Trends in genetics : TIG.

[52]  K. Birnbaum,et al.  Quantification of cell identity from single-cell gene expression profiles , 2015, Genome Biology.

[53]  Axel Visel,et al.  Genomic Perspectives of Transcriptional Regulation in Forebrain Development , 2015, Neuron.

[54]  Robert D. Finn,et al.  Rfam 12.0: updates to the RNA families database , 2014, Nucleic Acids Res..

[55]  D. Steffens,et al.  Basolateral amygdala volume and cell numbers in major depressive disorder: a postmortem stereological study , 2014, Brain Structure and Function.

[56]  Ross M. Fraser,et al.  Defining the role of common variation in the genomic and biological architecture of adult human height , 2014, Nature Genetics.

[57]  Paul Theodor Pyl,et al.  HTSeq—a Python framework to work with high-throughput sequencing data , 2014, bioRxiv.

[58]  Juan Carlos Fernández,et al.  Multiobjective evolutionary algorithms to identify highly autocorrelated areas: the case of spatial distribution in financially compromised farms , 2014, Ann. Oper. Res..

[59]  Yan Guo,et al.  Advanced Heat Map and Clustering Analysis Using Heatmap3 , 2014, BioMed research international.

[60]  Chuong B. Do,et al.  Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease , 2014, Nature Genetics.

[61]  Allan R. Jones,et al.  Transcriptional Landscape of the Prenatal Human Brain , 2014, Nature.

[62]  David R. O'Brien,et al.  Cell Type-Specific Expression Analysis to Identify Putative Cellular Mechanisms for Neurogenetic Disorders , 2014, The Journal of Neuroscience.

[63]  Mingfeng Li,et al.  Temporal Specification and Bilaterality of Human Neocortical Topographic Gene Expression , 2014, Neuron.

[64]  D. Spengler,et al.  Zac1 Regulates Cell Cycle Arrest in Neuronal Progenitors via Tcf4 , 2014, Molecular and Cellular Biology.

[65]  Daniel R. Zerbino,et al.  Ensembl 2014 , 2013, Nucleic Acids Res..

[66]  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.

[67]  Wei Shi,et al.  featureCounts: an efficient general purpose program for assigning sequence reads to genomic features , 2013, Bioinform..

[68]  S. Horvath,et al.  Integrative Functional Genomic Analyses Implicate Specific Molecular Pathways and Circuits in Autism , 2013, Cell.

[69]  Wei Niu,et al.  Coexpression Networks Implicate Human Midfetal Deep Cortical Projection Neurons in the Pathogenesis of Autism , 2013, Cell.

[70]  S. Hyman,et al.  Progress in the Genetics of Polygenic Brain Disorders: Significant New Challenges for Neurobiology , 2013, Neuron.

[71]  D. Geschwind,et al.  Cortical Evolution: Judge the Brain by Its Cover , 2013, Neuron.

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

[73]  Tanya M. Teslovich,et al.  Discovery and refinement of loci associated with lipid levels , 2013, Nature Genetics.

[74]  E. Levanon,et al.  Human housekeeping genes, revisited. , 2013, Trends in genetics : TIG.

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

[76]  L. Siever,et al.  Spatial and Temporal Mapping of De Novo Mutations in Schizophrenia to a Fetal Prefrontal Cortical Network , 2013, Cell.

[77]  P. Hof,et al.  Cerebellar granule cells are generated postnatally in humans , 2013, Brain Structure and Function.

[78]  W. Shi,et al.  The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote , 2013, Nucleic acids research.

[79]  Michael E. Greenberg,et al.  Activity-dependent neuronal signalling and autism spectrum disorder , 2013, Nature.

[80]  Mary Goldman,et al.  The UCSC Genome Browser database: extensions and updates 2013 , 2012, Nucleic Acids Res..

[81]  Kenneth Y Kwan,et al.  Transcriptional dysregulation of neocortical circuit assembly in ASD. , 2013, International review of neurobiology.

[82]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[83]  Brent S. Pedersen,et al.  Comb-p: software for combining, analyzing, grouping and correcting spatially correlated P-values , 2012, Bioinform..

[84]  Daniel J. Miller,et al.  Prolonged myelination in human neocortical evolution , 2012, Proceedings of the National Academy of Sciences.

[85]  Data production leads,et al.  An integrated encyclopedia of DNA elements in the human genome , 2012 .

[86]  Raymond K. Auerbach,et al.  An Integrated Encyclopedia of DNA Elements in the Human Genome , 2012, Nature.

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

[88]  W. Huber,et al.  Detecting differential usage of exons from RNA-seq data , 2012, Genome research.

[89]  Stephanie E. Vallee,et al.  Haploinsufficiency of SOX5 at 12p12.1 is associated with developmental delays with prominent language delay, behavior problems, and mild dysmorphic features , 2012, Human mutation.

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

[91]  K. Hansen,et al.  Removing technical variability in RNA-seq data using conditional quantile normalization , 2012, Biostatistics.

[92]  Sebastian D. Mackowiak,et al.  miRDeep2 accurately identifies known and hundreds of novel microRNA genes in seven animal clades , 2011, Nucleic acids research.

[93]  J. Leek,et al.  Temporal dynamics and genetic control of transcription in the human prefrontal cortex , 2011, Nature.

[94]  J. Kleinman,et al.  Spatiotemporal transcriptome of the human brain , 2011, Nature.

[95]  M. Salit,et al.  Synthetic Spike-in Standards for Rna-seq Experiments Material Supplemental Open Access License Commons Creative , 2022 .

[96]  A. Kriegstein,et al.  Development and Evolution of the Human Neocortex , 2011, Cell.

[97]  Mark Gerstein,et al.  RSEQtools: a modular framework to analyze RNA-Seq data using compact, anonymized data summaries , 2010, Bioinform..

[98]  Ana Kozomara,et al.  miRBase: integrating microRNA annotation and deep-sequencing data , 2010, Nucleic Acids Res..

[99]  Dave T. Gerrard,et al.  Gene expression divergence recapitulates the developmental hourglass model , 2010, Nature.

[100]  D. Tautz,et al.  A phylogenetically based transcriptome age index mirrors ontogenetic divergence patterns , 2010, Nature.

[101]  D. Altshuler,et al.  A map of human genome variation from population-scale sequencing , 2010, Nature.

[102]  C. W. Ragsdale,et al.  Molecular analysis of neocortical layer structure in the ferret , 2010, The Journal of comparative neurology.

[103]  H. Firth,et al.  Mutations in MEF2C from the 5q14.3q15 microdeletion syndrome region are a frequent cause of severe mental retardation and diminish MECP2 and CDKL5 expression , 2010, Human mutation.

[104]  W. Huber,et al.  which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. MAnorm: a robust model for quantitative comparison of ChIP-Seq data sets , 2011 .

[105]  Aaron R. Quinlan,et al.  BIOINFORMATICS APPLICATIONS NOTE , 2022 .

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

[107]  H. Firth,et al.  Mutations in MEF2C from the 5q14.3q15 microdeletion syndrome region are a frequent cause of severe mental retardation and diminish MECP2 and CDKL5 expression , 2010, Human mutation.

[108]  Chen Zeng,et al.  A clustering approach for identification of enriched domains from histone modification ChIP-Seq data , 2009, Bioinform..

[109]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[110]  D. Geschwind,et al.  Functional and Evolutionary Insights into Human Brain Development through Global Transcriptome Analysis , 2009, Neuron.

[111]  Steve Horvath,et al.  WGCNA: an R package for weighted correlation network analysis , 2008, BMC Bioinformatics.

[112]  T. Paus,et al.  Why do many psychiatric disorders emerge during adolescence? , 2008, Nature Reviews Neuroscience.

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

[114]  B. Williams,et al.  Mapping and quantifying mammalian transcriptomes by RNA-Seq , 2008, Nature Methods.

[115]  J. P. Hamilton,et al.  Amygdala volume in major depressive disorder: a meta-analysis of magnetic resonance imaging studies , 2008, Molecular Psychiatry.

[116]  Brad T. Sherman,et al.  Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources , 2008, Nature Protocols.

[117]  Geoffrey E. Hinton,et al.  Visualizing Data using t-SNE , 2008 .

[118]  Manuel A. R. Ferreira,et al.  PLINK: a tool set for whole-genome association and population-based linkage analyses. , 2007, American journal of human genetics.

[119]  Nathalie Boddaert,et al.  Mutations in TCF4, encoding a class I basic helix-loop-helix transcription factor, are responsible for Pitt-Hopkins syndrome, a severe epileptic encephalopathy associated with autonomic dysfunction. , 2007, American journal of human genetics.

[120]  Juliane Hoyer,et al.  Haploinsufficiency of TCF4 causes syndromal mental retardation with intermittent hyperventilation (Pitt-Hopkins syndrome). , 2007, American journal of human genetics.

[121]  Cheng Li,et al.  Adjusting batch effects in microarray expression data using empirical Bayes methods. , 2007, Biostatistics.

[122]  Wei Jiang,et al.  High-throughput DNA methylation profiling using universal bead arrays. , 2006, Genome research.

[123]  M. Ashburner,et al.  Gene Ontology: tool for the unification of biology , 2000, Nature Genetics.

[124]  Calmet Meteorological Model A User's Guide for the , 1999 .

[125]  P. Huttenlocher,et al.  Regional differences in synaptogenesis in human cerebral cortex , 1997, The Journal of comparative neurology.

[126]  T. Curran,et al.  A protein related to extracellular matrix proteins deleted in the mouse mutant reeler , 1995, Nature.

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

[128]  U. Francke,et al.  Phenotypic, cytogenetic, and molecular studies of three patients with constitutional deletions of chromosome 5 in the region of the gene for familial adenomatous polyposis. , 1992, American journal of human genetics.

[129]  B. Bernhardt,et al.  The economics of clinical genetics services. II. A time analysis of a medical genetics clinic. , 1987, American journal of human genetics.

[130]  K. Brodmann Vergleichende Lokalisationslehre der Großhirnrinde : in ihren Prinzipien dargestellt auf Grund des Zellenbaues , 1985 .

[131]  P. Huttenlocher Synaptic density in human frontal cortex - developmental changes and effects of aging. , 1979, Brain research.

[132]  R. Craig,et al.  Rate of circulation of the body fluid in adult Tenebrio molitor Linnaeus, Anasa tristis (de Goer), and Murgantia histrionica (Hahn). , 1951, Science.

[133]  S Paton,et al.  UNIVERSITY ADMINISTRATION AND UNIVERSITY IDEALS. , 1911, Science.

[134]  W. H. Dall Résultats du Voyage du S. Y. Belgica en 1897-9, sous the commandement de A. de Gerlache de Gomery , 1907 .