Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell ; 186(7): 1493-1511.e40, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001506

ABSTRACT

Understanding how genetic variants impact molecular phenotypes is a key goal of functional genomics, currently hindered by reliance on a single haploid reference genome. Here, we present the EN-TEx resource of 1,635 open-access datasets from four donors (∼30 tissues × âˆ¼15 assays). The datasets are mapped to matched, diploid genomes with long-read phasing and structural variants, instantiating a catalog of >1 million allele-specific loci. These loci exhibit coordinated activity along haplotypes and are less conserved than corresponding, non-allele-specific ones. Surprisingly, a deep-learning transformer model can predict the allele-specific activity based only on local nucleotide-sequence context, highlighting the importance of transcription-factor-binding motifs particularly sensitive to variants. Furthermore, combining EN-TEx with existing genome annotations reveals strong associations between allele-specific and GWAS loci. It also enables models for transferring known eQTLs to difficult-to-profile tissues (e.g., from skin to heart). Overall, EN-TEx provides rich data and generalizable models for more accurate personal functional genomics.


Subject(s)
Epigenome , Quantitative Trait Loci , Genome-Wide Association Study , Genomics , Phenotype , Polymorphism, Single Nucleotide
2.
Nat Immunol ; 24(8): 1382-1390, 2023 08.
Article in English | MEDLINE | ID: mdl-37500887

ABSTRACT

Microglia, the macrophages of the brain parenchyma, are key players in neurodegenerative diseases such as Alzheimer's disease. These cells adopt distinct transcriptional subtypes known as states. Understanding state function, especially in human microglia, has been elusive owing to a lack of tools to model and manipulate these cells. Here, we developed a platform for modeling human microglia transcriptional states in vitro. We found that exposure of human stem-cell-differentiated microglia to synaptosomes, myelin debris, apoptotic neurons or synthetic amyloid-beta fibrils generated transcriptional diversity that mapped to gene signatures identified in human brain microglia, including disease-associated microglia, a state enriched in neurodegenerative diseases. Using a new lentiviral approach, we demonstrated that the transcription factor MITF drives a disease-associated transcriptional signature and a highly phagocytic state. Together, these tools enable the manipulation and functional interrogation of human microglial states in both homeostatic and disease-relevant contexts.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Neurodegenerative Diseases , Humans , Microglia , Alzheimer Disease/genetics , Brain
3.
Cell ; 170(3): 522-533.e15, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753427

ABSTRACT

Genome-wide association studies (GWASs) implicate the PHACTR1 locus (6p24) in risk for five vascular diseases, including coronary artery disease, migraine headache, cervical artery dissection, fibromuscular dysplasia, and hypertension. Through genetic fine mapping, we prioritized rs9349379, a common SNP in the third intron of the PHACTR1 gene, as the putative causal variant. Epigenomic data from human tissue revealed an enhancer signature at rs9349379 exclusively in aorta, suggesting a regulatory function for this SNP in the vasculature. CRISPR-edited stem cell-derived endothelial cells demonstrate rs9349379 regulates expression of endothelin 1 (EDN1), a gene located 600 kb upstream of PHACTR1. The known physiologic effects of EDN1 on the vasculature may explain the pattern of risk for the five associated diseases. Overall, these data illustrate the integration of genetic, phenotypic, and epigenetic analysis to identify the biologic mechanism by which a common, non-coding variant can distally regulate a gene and contribute to the pathogenesis of multiple vascular diseases.


Subject(s)
Coronary Artery Disease/genetics , Endothelin-1/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Vascular Diseases/genetics , Acetylation , Cells, Cultured , Chromatin/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 6 , Endothelial Cells/cytology , Endothelin-1/blood , Epigenomics , Gene Editing , Gene Expression , Genome-Wide Association Study , Histones/metabolism , Humans , Muscle, Smooth, Vascular/cytology
4.
Cell ; 152(3): 642-54, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23333102

ABSTRACT

Differences in chromatin organization are key to the multiplicity of cell states that arise from a single genetic background, yet the landscapes of in vivo tissues remain largely uncharted. Here, we mapped chromatin genome-wide in a large and diverse collection of human tissues and stem cells. The maps yield unprecedented annotations of functional genomic elements and their regulation across developmental stages, lineages, and cellular environments. They also reveal global features of the epigenome, related to nuclear architecture, that also vary across cellular phenotypes. Specifically, developmental specification is accompanied by progressive chromatin restriction as the default state transitions from dynamic remodeling to generalized compaction. Exposure to serum in vitro triggers a distinct transition that involves de novo establishment of domains with features of constitutive heterochromatin. We describe how these global chromatin state transitions relate to chromosome and nuclear architecture, and discuss their implications for lineage fidelity, cellular senescence, and reprogramming.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Epigenesis, Genetic , Gene-Environment Interaction , Genome-Wide Association Study , Cell Nucleus , Cellular Senescence , Embryonic Stem Cells/metabolism , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/metabolism , Organ Specificity
5.
Cell ; 149(7): 1474-87, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726435

ABSTRACT

A large fraction of the mammalian genome is organized into inactive chromosomal domains along the nuclear lamina. The mechanism by which these lamina associated domains (LADs) are established remains to be elucidated. Using genomic repositioning assays, we show that LADs, spanning the developmentally regulated IgH and Cyp3a loci contain discrete DNA regions that associate chromatin with the nuclear lamina and repress gene activity in fibroblasts. Lamina interaction is established during mitosis and likely involves the localized recruitment of Lamin B during late anaphase. Fine-scale mapping of LADs reveals numerous lamina-associating sequences (LASs), which are enriched for a GAGA motif. This repeated motif directs lamina association and is bound by the transcriptional repressor cKrox, in a complex with HDAC3 and Lap2ß. Knockdown of cKrox or HDAC3 results in dissociation of LASs/LADs from the nuclear lamina. These results reveal a mechanism that couples nuclear compartmentalization of chromatin domains with the control of gene activity.


Subject(s)
Chromatin/genetics , DNA-Binding Proteins/metabolism , Gene Silencing , Mitosis , Nuclear Lamina/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , DNA/chemistry , Drosophila/metabolism , Histone Deacetylases/metabolism , Immunoglobulin Heavy Chains/genetics , Mice , NIH 3T3 Cells , Nuclear Envelope/metabolism , Transcription, Genetic
6.
Nature ; 593(7858): 238-243, 2021 05.
Article in English | MEDLINE | ID: mdl-33828297

ABSTRACT

Genome-wide association studies (GWAS) have identified thousands of noncoding loci that are associated with human diseases and complex traits, each of which could reveal insights into the mechanisms of disease1. Many of the underlying causal variants may affect enhancers2,3, but we lack accurate maps of enhancers and their target genes to interpret such variants. We recently developed the activity-by-contact (ABC) model to predict which enhancers regulate which genes and validated the model using CRISPR perturbations in several cell types4. Here we apply this ABC model to create enhancer-gene maps in 131 human cell types and tissues, and use these maps to interpret the functions of GWAS variants. Across 72 diseases and complex traits, ABC links 5,036 GWAS signals to 2,249 unique genes, including a class of 577 genes that appear to influence multiple phenotypes through variants in enhancers that act in different cell types. In inflammatory bowel disease (IBD), causal variants are enriched in predicted enhancers by more than 20-fold in particular cell types such as dendritic cells, and ABC achieves higher precision than other regulatory methods at connecting noncoding variants to target genes. These variant-to-function maps reveal an enhancer that contains an IBD risk variant and that regulates the expression of PPIF to alter the membrane potential of mitochondria in macrophages. Our study reveals principles of genome regulation, identifies genes that affect IBD and provides a resource and generalizable strategy to connect risk variants of common diseases to their molecular and cellular functions.


Subject(s)
Enhancer Elements, Genetic/genetics , Genetic Predisposition to Disease , Genetic Variation/genetics , Genome, Human/genetics , Genome-Wide Association Study , Inflammatory Bowel Diseases/genetics , Cell Line , Chromosomes, Human, Pair 10/genetics , Cyclophilins/genetics , Dendritic Cells , Female , Humans , Macrophages/metabolism , Male , Mitochondria/metabolism , Organ Specificity/genetics , Phenotype
7.
Cell ; 147(7): 1628-39, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22196736

ABSTRACT

Hundreds of chromatin regulators (CRs) control chromatin structure and function by catalyzing and binding histone modifications, yet the rules governing these key processes remain obscure. Here, we present a systematic approach to infer CR function. We developed ChIP-string, a meso-scale assay that combines chromatin immunoprecipitation with a signature readout of 487 representative loci. We applied ChIP-string to screen 145 antibodies, thereby identifying effective reagents, which we used to map the genome-wide binding of 29 CRs in two cell types. We found that specific combinations of CRs colocalize in characteristic patterns at distinct chromatin environments, at genes of coherent functions, and at distal regulatory elements. When comparing between cell types, CRs redistribute to different loci but maintain their modular and combinatorial associations. Our work provides a multiplex method that substantially enhances the ability to monitor CR binding, presents a large resource of CR maps, and reveals common principles for combinatorial CR function.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/metabolism , Genomics/methods , Histone Code , Chromatin/chemistry , Chromatin Assembly and Disassembly , Embryonic Stem Cells , Genome , Humans , K562 Cells
8.
Nature ; 583(7818): 699-710, 2020 07.
Article in English | MEDLINE | ID: mdl-32728249

ABSTRACT

The human and mouse genomes contain instructions that specify RNAs and proteins and govern the timing, magnitude, and cellular context of their production. To better delineate these elements, phase III of the Encyclopedia of DNA Elements (ENCODE) Project has expanded analysis of the cell and tissue repertoires of RNA transcription, chromatin structure and modification, DNA methylation, chromatin looping, and occupancy by transcription factors and RNA-binding proteins. Here we summarize these efforts, which have produced 5,992 new experimental datasets, including systematic determinations across mouse fetal development. All data are available through the ENCODE data portal (https://www.encodeproject.org), including phase II ENCODE1 and Roadmap Epigenomics2 data. We have developed a registry of 926,535 human and 339,815 mouse candidate cis-regulatory elements, covering 7.9 and 3.4% of their respective genomes, by integrating selected datatypes associated with gene regulation, and constructed a web-based server (SCREEN; http://screen.encodeproject.org) to provide flexible, user-defined access to this resource. Collectively, the ENCODE data and registry provide an expansive resource for the scientific community to build a better understanding of the organization and function of the human and mouse genomes.


Subject(s)
DNA/genetics , Databases, Genetic , Genome/genetics , Genomics , Molecular Sequence Annotation , Registries , Regulatory Sequences, Nucleic Acid/genetics , Animals , Chromatin/genetics , Chromatin/metabolism , DNA/chemistry , DNA Footprinting , DNA Methylation/genetics , DNA Replication Timing , Deoxyribonuclease I/metabolism , Genome, Human , Histones/metabolism , Humans , Mice , Mice, Transgenic , RNA-Binding Proteins/genetics , Transcription, Genetic/genetics , Transposases/metabolism
10.
Nature ; 518(7539): 337-43, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25363779

ABSTRACT

Genome-wide association studies have identified loci underlying human diseases, but the causal nucleotide changes and mechanisms remain largely unknown. Here we developed a fine-mapping algorithm to identify candidate causal variants for 21 autoimmune diseases from genotyping data. We integrated these predictions with transcription and cis-regulatory element annotations, derived by mapping RNA and chromatin in primary immune cells, including resting and stimulated CD4(+) T-cell subsets, regulatory T cells, CD8(+) T cells, B cells, and monocytes. We find that ∼90% of causal variants are non-coding, with ∼60% mapping to immune-cell enhancers, many of which gain histone acetylation and transcribe enhancer-associated RNA upon immune stimulation. Causal variants tend to occur near binding sites for master regulators of immune differentiation and stimulus-dependent gene activation, but only 10-20% directly alter recognizable transcription factor binding motifs. Rather, most non-coding risk variants, including those that alter gene expression, affect non-canonical sequence determinants not well-explained by current gene regulatory models.


Subject(s)
Autoimmune Diseases/genetics , Epigenesis, Genetic/genetics , Polymorphism, Single Nucleotide/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Base Sequence , Chromatin/genetics , Consensus Sequence/genetics , Enhancer Elements, Genetic/genetics , Epigenomics , Genome-Wide Association Study , Humans , Nucleotide Motifs , Organ Specificity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription Factors/metabolism
11.
Nature ; 518(7539): 317-30, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25693563

ABSTRACT

The reference human genome sequence set the stage for studies of genetic variation and its association with human disease, but epigenomic studies lack a similar reference. To address this need, the NIH Roadmap Epigenomics Consortium generated the largest collection so far of human epigenomes for primary cells and tissues. Here we describe the integrative analysis of 111 reference human epigenomes generated as part of the programme, profiled for histone modification patterns, DNA accessibility, DNA methylation and RNA expression. We establish global maps of regulatory elements, define regulatory modules of coordinated activity, and their likely activators and repressors. We show that disease- and trait-associated genetic variants are enriched in tissue-specific epigenomic marks, revealing biologically relevant cell types for diverse human traits, and providing a resource for interpreting the molecular basis of human disease. Our results demonstrate the central role of epigenomic information for understanding gene regulation, cellular differentiation and human disease.


Subject(s)
Epigenesis, Genetic/genetics , Epigenomics , Genome, Human/genetics , Base Sequence , Cell Lineage/genetics , Cells, Cultured , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromosomes, Human/chemistry , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Methylation , Datasets as Topic , Enhancer Elements, Genetic/genetics , Genetic Variation/genetics , Genome-Wide Association Study , Histones/metabolism , Humans , Organ Specificity/genetics , RNA/genetics , Reference Values
12.
Nature ; 512(7515): 449-52, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25164756

ABSTRACT

Genome function is dynamically regulated in part by chromatin, which consists of the histones, non-histone proteins and RNA molecules that package DNA. Studies in Caenorhabditis elegans and Drosophila melanogaster have contributed substantially to our understanding of molecular mechanisms of genome function in humans, and have revealed conservation of chromatin components and mechanisms. Nevertheless, the three organisms have markedly different genome sizes, chromosome architecture and gene organization. On human and fly chromosomes, for example, pericentric heterochromatin flanks single centromeres, whereas worm chromosomes have dispersed heterochromatin-like regions enriched in the distal chromosomal 'arms', and centromeres distributed along their lengths. To systematically investigate chromatin organization and associated gene regulation across species, we generated and analysed a large collection of genome-wide chromatin data sets from cell lines and developmental stages in worm, fly and human. Here we present over 800 new data sets from our ENCODE and modENCODE consortia, bringing the total to over 1,400. Comparison of combinatorial patterns of histone modifications, nuclear lamina-associated domains, organization of large-scale topological domains, chromatin environment at promoters and enhancers, nucleosome positioning, and DNA replication patterns reveals many conserved features of chromatin organization among the three organisms. We also find notable differences in the composition and locations of repressive chromatin. These data sets and analyses provide a rich resource for comparative and species-specific investigations of chromatin composition, organization and function.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Chromatin/genetics , Chromatin/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Animals , Cell Line , Centromere/genetics , Centromere/metabolism , Chromatin/chemistry , Chromatin Assembly and Disassembly/genetics , DNA Replication/genetics , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Heterochromatin/chemistry , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/chemistry , Histones/metabolism , Humans , Molecular Sequence Annotation , Nuclear Lamina/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics , Species Specificity
13.
Nature ; 473(7345): 43-9, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21441907

ABSTRACT

Chromatin profiling has emerged as a powerful means of genome annotation and detection of regulatory activity. The approach is especially well suited to the characterization of non-coding portions of the genome, which critically contribute to cellular phenotypes yet remain largely uncharted. Here we map nine chromatin marks across nine cell types to systematically characterize regulatory elements, their cell-type specificities and their functional interactions. Focusing on cell-type-specific patterns of promoters and enhancers, we define multicell activity profiles for chromatin state, gene expression, regulatory motif enrichment and regulator expression. We use correlations between these profiles to link enhancers to putative target genes, and predict the cell-type-specific activators and repressors that modulate them. The resulting annotations and regulatory predictions have implications for the interpretation of genome-wide association studies. Top-scoring disease single nucleotide polymorphisms are frequently positioned within enhancer elements specifically active in relevant cell types, and in some cases affect a motif instance for a predicted regulator, thus suggesting a mechanism for the association. Our study presents a general framework for deciphering cis-regulatory connections and their roles in disease.


Subject(s)
Cell Physiological Phenomena , Chromatin/genetics , Chromatin/metabolism , Chromosome Mapping , Binding Sites , Cell Line , Cell Line, Tumor , Cells, Cultured , Gene Expression Regulation , Genome, Human/genetics , Hep G2 Cells , Humans , Promoter Regions, Genetic/genetics , Reproducibility of Results , Transcription Factors/genetics
14.
Nat Methods ; 10(10): 949-55, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24076989

ABSTRACT

Epigenome-wide association studies (EWAS) hold promise for the detection of new regulatory mechanisms that may be susceptible to modification by environmental and lifestyle factors affecting susceptibility to disease. Epigenome-wide screening methods cover an increasing number of CpG sites, but the complexity of the data poses a challenge to separating robust signals from noise. Appropriate study design, a detailed a priori analysis plan and validation of results are essential to minimize the danger of false positive results and contribute to a unified approach. Epigenome-wide mapping studies in homogenous cell populations will inform our understanding of normal variation in the methylome that is not associated with disease or aging. Here we review concepts for conducting a stringent and powerful EWAS, including the choice of analyzed tissue, sources of variability and systematic biases, outline analytical solutions to EWAS-specific problems and highlight caveats in interpretation of data generated from samples with cellular heterogeneity.


Subject(s)
Epigenesis, Genetic , Genome-Wide Association Study/methods , Research Design , Animals , CpG Islands , DNA Methylation , Data Interpretation, Statistical , Genome-Wide Association Study/statistics & numerical data , Humans , Sequence Analysis, DNA
15.
Nucleic Acids Res ; 42(9): e74, 2014 May.
Article in English | MEDLINE | ID: mdl-24598259

ABSTRACT

In a chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq) experiment, an important consideration in experimental design is the minimum number of sequenced reads required to obtain statistically significant results. We present an extensive evaluation of the impact of sequencing depth on identification of enriched regions for key histone modifications (H3K4me3, H3K36me3, H3K27me3 and H3K9me2/me3) using deep-sequenced datasets in human and fly. We propose to define sufficient sequencing depth as the number of reads at which detected enrichment regions increase <1% for an additional million reads. Although the required depth depends on the nature of the mark and the state of the cell in each experiment, we observe that sufficient depth is often reached at <20 million reads for fly. For human, there are no clear saturation points for the examined datasets, but our analysis suggests 40-50 million reads as a practical minimum for most marks. We also devise a mathematical model to estimate the sufficient depth and total genomic coverage of a mark. Lastly, we find that the five algorithms tested do not agree well for broad enrichment profiles, especially at lower depths. Our findings suggest that sufficient sequencing depth and an appropriate peak-calling algorithm are essential for ensuring robustness of conclusions derived from ChIP-seq data.


Subject(s)
High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA , Algorithms , Animals , Chromatin Immunoprecipitation , Drosophila melanogaster/genetics , Genome, Human , Genome, Insect , Genomic Library , Histones/metabolism , Humans , Models, Genetic , Protein Processing, Post-Translational
16.
PLoS Genet ; 7(12): e1002389, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22174693

ABSTRACT

DNA methylation plays an important role in development and disease. The primary sites of DNA methylation in vertebrates are cytosines in the CpG dinucleotide context, which account for roughly three quarters of the total DNA methylation content in human and mouse cells. While the genomic distribution, inter-individual stability, and functional role of CpG methylation are reasonably well understood, little is known about DNA methylation targeting CpA, CpT, and CpC (non-CpG) dinucleotides. Here we report a comprehensive analysis of non-CpG methylation in 76 genome-scale DNA methylation maps across pluripotent and differentiated human cell types. We confirm non-CpG methylation to be predominantly present in pluripotent cell types and observe a decrease upon differentiation and near complete absence in various somatic cell types. Although no function has been assigned to it in pluripotency, our data highlight that non-CpG methylation patterns reappear upon iPS cell reprogramming. Intriguingly, the patterns are highly variable and show little conservation between different pluripotent cell lines. We find a strong correlation of non-CpG methylation and DNMT3 expression levels while showing statistical independence of non-CpG methylation from pluripotency associated gene expression. In line with these findings, we show that knockdown of DNMTA and DNMT3B in hESCs results in a global reduction of non-CpG methylation. Finally, non-CpG methylation appears to be spatially correlated with CpG methylation. In summary these results contribute further to our understanding of cytosine methylation patterns in human cells using a large representative sample set.


Subject(s)
CpG Islands/genetics , Cytosine/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , Animals , Cell Differentiation , Cell Line , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Gene Expression , Gene Knockdown Techniques , Genome, Human , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Pluripotent Stem Cells/metabolism , RNA, Small Interfering/genetics , DNA Methyltransferase 3B
17.
Nat Commun ; 15(1): 2106, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38453887

ABSTRACT

In all terrestrial vertebrates, the parathyroid glands are critical regulators of calcium homeostasis and the sole source of parathyroid hormone (PTH). Hyperparathyroidism and hypoparathyroidism are clinically important disorders affecting multiple organs. However, our knowledge regarding regulatory mechanisms governing the parathyroids has remained limited. Here, we present the comprehensive maps of the chromatin landscape of the human parathyroid glands, identifying active regulatory elements and chromatin interactions. These data allow us to define regulatory circuits and previously unidentified genes that play crucial roles in parathyroid biology. We experimentally validate candidate parathyroid-specific enhancers and demonstrate their integration with GWAS SNPs for parathyroid-related diseases and traits. For instance, we observe reduced activity of a parathyroid-specific enhancer of the Calcium Sensing Receptor gene, which contains a risk allele associated with higher PTH levels compared to the wildtype allele. Our datasets provide a valuable resource for unraveling the mechanisms governing parathyroid gland regulation in health and disease.


Subject(s)
Calcium , Parathyroid Glands , Animals , Humans , Calcium/metabolism , Parathyroid Glands/metabolism , Parathyroid Hormone/genetics , Parathyroid Hormone/metabolism , Chromatin/genetics , Epigenesis, Genetic
18.
bioRxiv ; 2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37034577

ABSTRACT

Cis-regulatory elements control gene expression and are dynamic in their structure, reflecting changes to the composition of diverse effector proteins over time1-3. Here we sought to connect the structural changes at cis-regulatory elements to alterations in cellular fate and function. To do this we developed PRINT, a computational method that uses deep learning to correct sequence bias in chromatin accessibility data and identifies multi-scale footprints of DNA-protein interactions. We find that multi-scale footprints enable more accurate inference of TF and nucleosome binding. Using PRINT with single-cell multi-omics, we discover wide-spread changes to the structure and function of candidate cis-regulatory elements (cCREs) across hematopoiesis, wherein nucleosomes slide, expose DNA for TF binding, and promote gene expression. Activity segmentation using the co-variance across cell states identifies "sub-cCREs" as modular cCRE subunits of regulatory DNA. We apply this single-cell and PRINT approach to characterize the age-associated alterations to cCREs within hematopoietic stem cells (HSCs). Remarkably, we find a spectrum of aging alterations among HSCs corresponding to a global gain of sub-cCRE activity while preserving cCRE accessibility. Collectively, we reveal the functional importance of cCRE structure across cell states, highlighting changes to gene regulation at single-cell and single-base-pair resolution.

19.
J Invest Dermatol ; 141(5): 1207-1218, 2021 05.
Article in English | MEDLINE | ID: mdl-33212152

ABSTRACT

Basal cell carcinoma (BCC) and squamous cell carcinoma (SCC) are both derived from epidermal keratinocytes but are phenotypically diverse. To improve the understanding of keratinocyte carcinogenesis, it is critical to understand epigenetic alterations, especially those that govern gene expression. We examined changes to the enhancer-associated histone acetylation mark H3K27ac by mapping matched tumor-normal pairs from 11 patients (five with BCC and six with SCC) undergoing Mohs surgery. Our analysis uncovered cancer-specific enhancers on the basis of differential H3K27ac peaks between matched tumor-normal pairs. We also uncovered biological pathways potentially altered in keratinocyte carcinoma, including enriched epidermal development and Wnt signaling pathways enriched in BCCs and enriched immune response and cell activation pathways in SCCs. We also observed enrichment of transcription factors that implicated SMAD and JDP2 in BCC pathogenesis and FOXP1 in SCC pathogenesis. On the basis of these findings, we prioritized three loci with putative regulation events (FGFR2 enhancer in BCC, intragenic regulation of FOXP1 in SCC, and WNT5A promoter in both subtypes) and validated our findings with published gene expression data. Our findings highlight unique and shared epigenetic alterations in histone modifications and potential regulators for BCCs and SCCs that likely impact the divergent oncogenic pathways, paving the way for targeted drug discoveries.


Subject(s)
Carcinoma, Basal Cell/genetics , Carcinoma, Squamous Cell/genetics , Epigenesis, Genetic , Skin Neoplasms/genetics , Aged , Aged, 80 and over , Enhancer Elements, Genetic , Female , Humans , Male , Middle Aged , Receptor, Fibroblast Growth Factor, Type 2/genetics , Transcription, Genetic , Transcriptome , Wnt-5a Protein/genetics
20.
Nat Commun ; 11(1): 3697, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32728101

ABSTRACT

As the number of genomics datasets grows rapidly, sample mislabeling has become a high stakes issue. We present CrosscheckFingerprints (Crosscheck), a tool for quantifying sample-relatedness and detecting incorrectly paired sequencing datasets from different donors. Crosscheck outperforms similar methods and is effective even when data are sparse or from different assays. Application of Crosscheck to 8851 ENCODE ChIP-, RNA-, and DNase-seq datasets enabled us to identify and correct dozens of mislabeled samples and ambiguous metadata annotations, representing ~1% of ENCODE datasets.


Subject(s)
High-Throughput Nucleotide Sequencing , Linkage Disequilibrium/genetics , Databases, Nucleic Acid , Genotype , HEK293 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , K562 Cells , Lod Score , Molecular Sequence Annotation
SELECTION OF CITATIONS
SEARCH DETAIL