Brain cell type–specific enhancer–promoter interactome maps and disease-risk association

Linking enhancers to disease Enhancers are genomic regions that regulate gene expression, sometimes in a cell-dependent manner. However, most of our knowledge of human brain cell–type enhancers derives from studies of bulk human brain tissue. Nott et al. examined chromatin and promoter activity in cell nuclei isolated from human brains. Genetic variants associated with brain traits and disease showed cell-specific patterns of enhancer enrichment. These data indicate that Alzheimer's disease is regulated by genetic variants within microglial cells, whereas psychiatric diseases tend to affect neurons. Science, this issue p. 1134 Cell type–specific regulatory elements in the human brain reveal noncoding variants associated with neurological diseases. Noncoding genetic variation is a major driver of phenotypic diversity, but functional interpretation is challenging. To better understand common genetic variation associated with brain diseases, we defined noncoding regulatory regions for major cell types of the human brain. Whereas psychiatric disorders were primarily associated with variants in transcriptional enhancers and promoters in neurons, sporadic Alzheimer’s disease (AD) variants were largely confined to microglia enhancers. Interactome maps connecting disease-risk variants in cell-type–specific enhancers to promoters revealed an extended microglia gene network in AD. Deletion of a microglia-specific enhancer harboring AD-risk variants ablated BIN1 expression in microglia, but not in neurons or astrocytes. These findings revise and expand the list of genes likely to be influenced by noncoding variants in AD and suggest the probable cell types in which they function.

[1]  Eun Yong Kang,et al.  Identifying Causal Variants at Loci with Multiple Signals of Association , 2014, Genetics.

[2]  A. Auton,et al.  Genome-wide analysis of insomnia in 1,331,010 individuals identifies new risk loci and functional pathways , 2019, Nature Genetics.

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

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

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

[6]  Tuan Leng Tay,et al.  Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation , 2019, Science.

[7]  Cole Trapnell,et al.  Ultrafast and memory-efficient alignment of short DNA sequences to the human genome , 2009, Genome Biology.

[8]  Sonja W. Scholz,et al.  Parkinson’s disease genetics: identifying novel risk loci, providing causal insights and improving estimates of heritable risk , 2018, bioRxiv.

[9]  R. Young,et al.  Super-Enhancers in the Control of Cell Identity and Disease , 2013, Cell.

[10]  Kornel Labun,et al.  CHOPCHOP v2: a web tool for the next generation of CRISPR genome engineering , 2016, Nucleic Acids Res..

[11]  Stuart J. Ritchie,et al.  Study of 300,486 individuals identifies 148 independent genetic loci influencing general cognitive function , 2018, Nature Communications.

[12]  Niyazi Ari,et al.  Matplotlib in python , 2014, 2014 11th International Conference on Electronics, Computer and Computation (ICECCO).

[13]  Bing Ren,et al.  MAPS: Model-based analysis of long-range chromatin interactions from PLAC-seq and HiChIP experiments , 2018, bioRxiv.

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

[15]  I. Amit,et al.  A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease , 2017, Cell.

[16]  Christian Gieger,et al.  Genome-wide mega-analysis identifies 16 loci and highlights diverse biological mechanisms in the common epilepsies , 2018, Nature Communications.

[17]  Toby J. Gibson,et al.  KEPE—a motif frequently superimposed on sumoylation sites in metazoan chromatin proteins and transcription factors , 2008, Bioinform..

[18]  Annelot M. Dekker,et al.  Genome-wide association analyses identify new risk variants and the genetic architecture of amyotrophic lateral sclerosis , 2016, Nature Genetics.

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

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

[21]  Howard Y. Chang,et al.  HiChIP: efficient and sensitive analysis of protein-directed genome architecture , 2016, Nature Methods.

[22]  Olga Tanaseichuk,et al.  Metascape provides a biologist-oriented resource for the analysis of systems-level datasets , 2019, Nature Communications.

[23]  M. Hill,et al.  Enrichment of schizophrenia heritability in both neuronal and glia cell regulatory elements , 2018, Translational Psychiatry.

[24]  M. Daly,et al.  An Atlas of Genetic Correlations across Human Diseases and Traits , 2015, Nature Genetics.

[25]  Jonathan P. Beauchamp,et al.  Genome-wide association analyses of risk tolerance and risky behaviors in over 1 million individuals identify hundreds of loci and shared genetic influences , 2019, Nature Genetics.

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

[27]  A. Quinlan BEDTools: The Swiss‐Army Tool for Genome Feature Analysis , 2014, Current protocols in bioinformatics.

[28]  Jianjun Luo,et al.  BioCircos.js: an interactive Circos JavaScript library for biological data visualization on web applications , 2016, Bioinform..

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

[30]  Manolis Kellis,et al.  Single-cell transcriptomic analysis of Alzheimer’s disease , 2019, Nature.

[31]  Shane J. Neph,et al.  Systematic Localization of Common Disease-Associated Variation in Regulatory DNA , 2012, Science.

[32]  M. Pirinen,et al.  Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis , 2013, Nature Genetics.

[33]  F. Gage,et al.  Generation of multiciliated cells in functional airway epithelia from human induced pluripotent stem cells , 2014, Proceedings of the National Academy of Sciences.

[34]  E. Eskin,et al.  Integrating Functional Data to Prioritize Causal Variants in Statistical Fine-Mapping Studies , 2014, PLoS genetics.

[35]  Genetic,et al.  Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing , 2019, Nature Genetics.

[36]  Galt P. Barber,et al.  BigWig and BigBed: enabling browsing of large distributed datasets , 2010, Bioinform..

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

[38]  C. Glass,et al.  Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.

[39]  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.

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

[41]  C. Glass,et al.  The selection and function of cell type-specific enhancers , 2015, Nature Reviews Molecular Cell Biology.

[42]  R. Young,et al.  Histone H3K27ac separates active from poised enhancers and predicts developmental state , 2010, Proceedings of the National Academy of Sciences.

[43]  C. Glass,et al.  Analysis of Genetically Diverse Macrophages Reveals Local and Domain-wide Mechanisms that Control Transcription Factor Binding and Function , 2018, Cell.

[44]  Baptiste N. Jaeger,et al.  Pathological priming causes developmental gene network heterochronicity in autism patient-derived neurons , 2019, Nature Neuroscience.

[45]  Tom H. Pringle,et al.  The human genome browser at UCSC. , 2002, Genome research.

[46]  A. Chen-Plotkin,et al.  The Post-GWAS Era: From Association to Function. , 2018, American journal of human genetics.

[47]  T. Hughes,et al.  The Human Transcription Factors , 2018, Cell.

[48]  Galina A. Erikson,et al.  Stem Cell Reports Resource Differentiation of Inflammation-Responsive Astrocytes fromGlial Progenitors Generated from Human Induced Pluripotent Stem Cells , 2017 .

[49]  Howard Y. Chang,et al.  ATAC-see reveals the accessible genome by transposase-mediated imaging and sequencing , 2016, Nature Methods.

[50]  E. Chang,et al.  Purification and Characterization of Progenitor and Mature Human Astrocytes Reveals Transcriptional and Functional Differences with Mouse , 2016, Neuron.

[51]  K. Estrada,et al.  BIN1 favors the spreading of Tau via extracellular vesicles , 2019, Scientific Reports.

[52]  Nathaniel D. Heintzman,et al.  Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome , 2007, Nature Genetics.

[53]  M. Hill,et al.  Genetic risk for Alzheimer’s disease is concentrated in specific macrophage and microglial transcriptional networks , 2018, Genome Medicine.

[54]  K. Hao,et al.  A common haplotype lowers PU.1 expression in myeloid cells and delays onset of Alzheimer's disease , 2017, Nature Neuroscience.

[55]  J. Thomson,et al.  Embryonic stem cell lines derived from human blastocysts. , 1998, Science.

[56]  Baptiste N. Jaeger,et al.  An environment-dependent transcriptional network specifies human microglia identity , 2017, Science.

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

[58]  Miao Yu,et al.  Mapping of long-range chromatin interactions by proximity ligation-assisted ChIP-seq , 2016, Cell Research.

[59]  Integration of Alzheimer’s disease genetics and myeloid genomics reveals novel disease risk mechanisms , 2019 .

[60]  John P. Rice,et al.  Identification of common genetic risk variants for autism spectrum disorder , 2019, Nature Genetics.

[61]  Peter J. Bickel,et al.  Measuring reproducibility of high-throughput experiments , 2011, 1110.4705.

[62]  M. Blurton-Jones,et al.  Development and validation of a simplified method to generate human microglia from pluripotent stem cells , 2018, Molecular Neurodegeneration.

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

[64]  Anastasia G. Efthymiou,et al.  Integration of Alzheimer’s disease genetics and myeloid genomics reveals novel disease risk mechanisms , 2019, bioRxiv.

[65]  Sonja W. Scholz,et al.  Expanding Parkinson’s disease genetics: novel risk loci, genomic context, causal insights and heritable risk , 2018 .

[66]  Timothy J. Hohman,et al.  Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk , 2019, Nature Genetics.

[67]  S. Linnarsson,et al.  Meta-analysis of genome-wide association studies for neuroticism in 449,484 individuals identifies novel genetic loci and pathways , 2018, Nature Genetics.