Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 17445, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060836

ABSTRACT

CHARGE syndrome, a rare multiple congenital anomaly condition, is caused by haploinsufficiency of the chromatin remodeling protein gene CHD7 (Chromodomain helicase DNA binding protein 7). Brain abnormalities and intellectual disability are commonly observed in individuals with CHARGE, and neuronal differentiation is reduced in CHARGE patient-derived iPSCs and conditional knockout mouse brains. However, the mechanisms of CHD7 function in nervous system development are not well understood. In this study, we asked whether CHD7 promotes gene transcription in neural progenitor cells via changes in chromatin accessibility. We used Chd7 null embryonic stem cells (ESCs) derived from Chd7 mutant mouse blastocysts as a tool to investigate roles of CHD7 in neuronal and glial differentiation. Loss of Chd7 significantly reduced neuronal and glial differentiation. Sholl analysis showed that loss of Chd7 impaired neuronal complexity and neurite length in differentiated neurons. Genome-wide studies demonstrated that loss of Chd7 leads to modified chromatin accessibility (ATAC-seq) and differential nascent expression (Bru-Seq) of neural-specific genes. These results suggest that CHD7 acts preferentially to alter chromatin accessibility of key genes during the transition of NPCs to neurons to promote differentiation. Our results form a basis for understanding the cell stage-specific roles for CHD7-mediated chromatin remodeling during cell lineage acquisition.


Subject(s)
Chromatin/chemistry , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Neural Stem Cells/cytology , Neurons/cytology , Animals , Blastocyst/metabolism , Cell Differentiation , Enhancer Elements, Genetic , Epigenesis, Genetic , Gene Expression Profiling , Mice , Mice, Knockout , Transcription Factors/metabolism
2.
J Clin Invest ; 129(10): 4377-4392, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31498151

ABSTRACT

Despite progress in intensification of therapy, outcomes for patients with metastatic osteosarcoma (OS) have not improved in thirty years. We developed a system that enabled preclinical screening of compounds against metastatic OS cells in the context of the native lung microenvironment. Using this strategy to screen a library of epigenetically targeted compounds, we identified inhibitors of CDK12 to be most effective, reducing OS cell outgrowth in the lung by more than 90% at submicromolar doses. We found that knockout of CDK12 in an in vivo model of lung metastasis significantly decreased the ability of OS to colonize the lung. CDK12 inhibition led to defects in transcription elongation in a gene length- and expression-dependent manner. These effects were accompanied by defects in RNA processing and altered the expression of genes involved in transcription regulation and the DNA damage response. We further identified OS models that differ in their sensitivity to CDK12 inhibition in the lung and provided evidence that upregulated MYC levels may mediate these differences. Our studies provided a framework for rapid preclinical testing of compounds with antimetastatic activity and highlighted CDK12 as a potential therapeutic target in OS.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Osteosarcoma/enzymology , Osteosarcoma/secondary , Animals , Cell Line, Tumor , Cyclin-Dependent Kinases/genetics , Drug Screening Assays, Antitumor , Female , Gene Knockout Techniques , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice , Mice, SCID , Osteosarcoma/genetics , Protein Kinase Inhibitors/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology
3.
Elife ; 82019 02 13.
Article in English | MEDLINE | ID: mdl-30759065

ABSTRACT

Commonly-mutated genes have been found for many cancers, but less is known about mutations in cis-regulatory elements. We leverage gains in tumor-specific enhancer activity, coupled with allele-biased mutation detection from H3K27ac ChIP-seq data, to pinpoint potential enhancer-activating mutations in colorectal cancer (CRC). Analysis of a genetically-diverse cohort of CRC specimens revealed that microsatellite instable (MSI) samples have a high indel rate within active enhancers. Enhancers with indels show evidence of positive selection, increased target gene expression, and a subset is highly recurrent. The indels affect short homopolymer tracts of A/T and increase affinity for FOX transcription factors. We further demonstrate that signature mismatch-repair (MMR) mutations activate enhancers using a xenograft tumor metastasis model, where mutations are induced naturally via CRISPR/Cas9 inactivation of MLH1 prior to tumor cell injection. Our results suggest that MMR signature mutations activate enhancers in CRC tumor epigenomes to provide a selective advantage.


Subject(s)
Colorectal Neoplasms/genetics , DNA Mismatch Repair/genetics , Enhancer Elements, Genetic/genetics , Epigenome , Mutation/genetics , Acetylation , Animals , Base Sequence , Cell Line, Tumor , Gene Expression Regulation , Histones/metabolism , Humans , INDEL Mutation/genetics , Lysine/metabolism , Mice , Microsatellite Instability , Nucleotide Motifs/genetics , Phenotype , Selection, Genetic , Transcription Factors/metabolism
4.
Nat Med ; 24(9): 1469-1480, 2018 09.
Article in English | MEDLINE | ID: mdl-30038216

ABSTRACT

The degree of intrinsic and interpatient phenotypic heterogeneity and its role in tumor evolution is poorly understood. Phenotypic drifts can be transmitted via inheritable transcriptional programs. Cell-type specific transcription is maintained through the activation of epigenetically defined regulatory regions including promoters and enhancers. Here we have annotated the epigenome of 47 primary and metastatic estrogen-receptor (ERα)-positive breast cancer clinical specimens and inferred phenotypic heterogeneity from the regulatory landscape, identifying key regulatory elements commonly shared across patients. Shared regions contain a unique set of regulatory information including the motif for transcription factor YY1. We identify YY1 as a critical determinant of ERα transcriptional activity promoting tumor growth in most luminal patients. YY1 also contributes to the expression of genes mediating resistance to endocrine treatment. Finally, we used H3K27ac levels at active enhancer elements as a surrogate of intra-tumor phenotypic heterogeneity to track the expansion and contraction of phenotypic subpopulations throughout breast cancer progression. By tracking the clonality of SLC9A3R1-positive cells, a bona fide YY1-ERα-regulated gene, we show that endocrine therapies select for phenotypic clones under-represented at diagnosis. Collectively, our data show that epigenetic mechanisms significantly contribute to phenotypic heterogeneity and evolution in systemically treated breast cancer patients.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Clonal Evolution , Enhancer Elements, Genetic/genetics , Cell Line, Tumor , Clone Cells , Epigenesis, Genetic/drug effects , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Female , Humans , MCF-7 Cells , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , Polymorphism, Single Nucleotide/genetics , Protein Binding/drug effects , Risk Factors , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Transcription, Genetic/drug effects , YY1 Transcription Factor/metabolism
5.
Sci Rep ; 8(1): 7324, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29743621

ABSTRACT

Long non-coding RNAs (lncRNAs) are frequently dysregulated in many human cancers. We sought to identify candidate oncogenic lncRNAs in human colon tumors by utilizing RNA sequencing data from 22 colon tumors and 22 adjacent normal colon samples from The Cancer Genome Atlas (TCGA). The analysis led to the identification of ~200 differentially expressed lncRNAs. Validation in an independent cohort of normal colon and patient-derived colon cancer cell lines identified a novel lncRNA, lincDUSP, as a potential candidate oncogene. Knockdown of lincDUSP in patient-derived colon tumor cell lines resulted in significantly decreased cell proliferation and clonogenic potential, and increased susceptibility to apoptosis. The knockdown of lincDUSP affects the expression of ~800 genes, and NCI pathway analysis showed enrichment of DNA damage response and cell cycle control pathways. Further, identification of lincDUSP chromatin occupancy sites by ChIRP-Seq demonstrated association with genes involved in the replication-associated DNA damage response and cell cycle control. Consistent with these findings, lincDUSP knockdown in colon tumor cell lines increased both the accumulation of cells in early S-phase and γH2AX foci formation, indicating increased DNA damage response induction. Taken together, these results demonstrate a key role of lincDUSP in the regulation of important pathways in colon cancer.


Subject(s)
Apoptosis/genetics , Cell Cycle/genetics , Colonic Neoplasms/pathology , RNA, Long Noncoding/genetics , Up-Regulation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colonic Neoplasms/genetics , DNA Damage/genetics , Gene Knockdown Techniques , Genomics , Humans , RNA, Long Noncoding/metabolism
7.
Nat Med ; 24(2): 176-185, 2018 02.
Article in English | MEDLINE | ID: mdl-29334376

ABSTRACT

Metastasis results from a complex set of traits acquired by tumor cells, distinct from those necessary for tumorigenesis. Here, we investigate the contribution of enhancer elements to the metastatic phenotype of osteosarcoma. Through epigenomic profiling, we identify substantial differences in enhancer activity between primary and metastatic human tumors and between near isogenic pairs of highly lung metastatic and nonmetastatic osteosarcoma cell lines. We term these regions metastatic variant enhancer loci (Met-VELs). Met-VELs drive coordinated waves of gene expression during metastatic colonization of the lung. Met-VELs cluster nonrandomly in the genome, indicating that activity of these enhancers and expression of their associated gene targets are positively selected. As evidence of this causal association, osteosarcoma lung metastasis is inhibited by global interruptions of Met-VEL-associated gene expression via pharmacologic BET inhibition, by knockdown of AP-1 transcription factors that occupy Met-VELs, and by knockdown or functional inhibition of individual genes activated by Met-VELs, such as that encoding coagulation factor III/tissue factor (F3). We further show that genetic deletion of a single Met-VEL at the F3 locus blocks metastatic cell outgrowth in the lung. These findings indicate that Met-VELs and the genes they regulate play a functional role in metastasis and may be suitable targets for antimetastatic therapies.


Subject(s)
Carcinogenesis/genetics , Enhancer Elements, Genetic/genetics , Lung Neoplasms/genetics , Osteosarcoma/genetics , Cell Line, Tumor , Epigenomics , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Neoplasm Metastasis/genetics , Osteosarcoma/pathology , Proteins/antagonists & inhibitors , Proteins/genetics , Selection, Genetic , Thromboplastin/genetics , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/genetics , Tumor Microenvironment/genetics
8.
Nature ; 553(7686): 101-105, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29258295

ABSTRACT

Genomic sequencing has driven precision-based oncology therapy; however, the genetic drivers of many malignancies remain unknown or non-targetable, so alternative approaches to the identification of therapeutic leads are necessary. Ependymomas are chemotherapy-resistant brain tumours, which, despite genomic sequencing, lack effective molecular targets. Intracranial ependymomas are segregated on the basis of anatomical location (supratentorial region or posterior fossa) and further divided into distinct molecular subgroups that reflect differences in the age of onset, gender predominance and response to therapy. The most common and aggressive subgroup, posterior fossa ependymoma group A (PF-EPN-A), occurs in young children and appears to lack recurrent somatic mutations. Conversely, posterior fossa ependymoma group B (PF-EPN-B) tumours display frequent large-scale copy number gains and losses but have favourable clinical outcomes. More than 70% of supratentorial ependymomas are defined by highly recurrent gene fusions in the NF-κB subunit gene RELA (ST-EPN-RELA), and a smaller number involve fusion of the gene encoding the transcriptional activator YAP1 (ST-EPN-YAP1). Subependymomas, a distinct histologic variant, can also be found within the supratetorial and posterior fossa compartments, and account for the majority of tumours in the molecular subgroups ST-EPN-SE and PF-EPN-SE. Here we describe mapping of active chromatin landscapes in 42 primary ependymomas in two non-overlapping primary ependymoma cohorts, with the goal of identifying essential super-enhancer-associated genes on which tumour cells depend. Enhancer regions revealed putative oncogenes, molecular targets and pathways; inhibition of these targets with small molecule inhibitors or short hairpin RNA diminished the proliferation of patient-derived neurospheres and increased survival in mouse models of ependymomas. Through profiling of transcriptional enhancers, our study provides a framework for target and drug discovery in other cancers that lack known genetic drivers and are therefore difficult to treat.


Subject(s)
Enhancer Elements, Genetic/genetics , Ependymoma/drug therapy , Ependymoma/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks/genetics , Molecular Targeted Therapy , Oncogenes/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Ependymoma/classification , Ependymoma/pathology , Female , Humans , Mice , Precision Medicine , RNA Interference , Xenograft Model Antitumor Assays
9.
G3 (Bethesda) ; 8(2): 491-503, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29223978

ABSTRACT

A hallmark of craniofacial development is the differentiation of multiple cell lineages in close proximity to one another. The mouse skull bones and overlying dermis are derived from the cranial mesenchyme (CM). Cell fate selection of the embryonic cranial bone and dermis in the CM requires Wnt/ß-catenin signaling, and loss of ß-catenin leads to an ectopic chondrogenic cell fate switch. The mechanism by which Wnt/ß-catenin activity suppresses the cartilage fate is unclear. Upon conditional deletion of ß-catenin in the CM, several key determinants of the cartilage differentiation program, including Sox9, become differentially expressed. Many of these differentially expressed genes are known targets of the Polycomb Repressive Complex 2 (PRC2). Thus, we hypothesized that PRC2 is required for Wnt/ß-catenin-mediated repression of chondrogenesis in the embryonic CM. We find that ß-catenin can physically interact with PRC2 components in the CM in vivo However, upon genetic deletion of Enhancer of Zeste homolog 2 (EZH2), the catalytic component of PRC2, chondrogenesis remains repressed and the bone and dermis cell fate is preserved in the CM. Furthermore, loss of ß-catenin does not alter either the H3K27me3 enrichment levels genome-wide or on cartilage differentiation determinants, including Sox9 Our results indicate that EZH2 is not required to repress chondrogenesis in the CM downstream of Wnt/ß-catenin signaling.


Subject(s)
Chondrogenesis/genetics , Mesoderm/metabolism , Polycomb Repressive Complex 2/genetics , Skull/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Animals , Cartilage/cytology , Cartilage/embryology , Cartilage/metabolism , Cell Differentiation/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Mesoderm/cytology , Mesoderm/embryology , Mice, Knockout , Mice, Transgenic , Polycomb Repressive Complex 2/metabolism , Skull/cytology , Skull/embryology , beta Catenin/metabolism
10.
Nat Commun ; 8: 14400, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28169291

ABSTRACT

In addition to mutations in genes, aberrant enhancer element activity at non-coding regions of the genome is a key driver of tumorigenesis. Here, we perform epigenomic enhancer profiling of a cohort of more than forty genetically diverse human colorectal cancer (CRC) specimens. Using normal colonic crypt epithelium as a comparator, we identify enhancers with recurrently gained or lost activity across CRC specimens. Of the enhancers highly recurrently activated in CRC, most are constituents of super enhancers, are occupied by AP-1 and cohesin complex members, and originate from primed chromatin. Many activate known oncogenes, and CRC growth can be mitigated through pharmacologic inhibition or genome editing of these loci. Nearly half of all GWAS CRC risk loci co-localize to recurrently activated enhancers. These findings indicate that the CRC epigenome is defined by highly recurrent epigenetic alterations at enhancers which activate a common, aberrant transcriptional programme critical for CRC growth and survival.


Subject(s)
Colorectal Neoplasms/genetics , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic , Genetic Loci/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/surgery , Datasets as Topic , Epigenomics/methods , Female , Humans , Mice , Mice, Nude , Mutation , Tissue Array Analysis , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(33): 53230-53244, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27449296

ABSTRACT

Approximately, 25-30% of early-stage breast tumors are classified at the molecular level as HER2-positive, which is an aggressive subtype of breast cancer. Amplification of the HER2 gene in these tumors results in a substantial increase in HER2 mRNA levels, and consequently, HER2 protein levels. HER2, a transmembrane receptor tyrosine kinase (RTK), is targeted therapeutically by a monoclonal antibody, trastuzumab (Tz), which has dramatically improved the prognosis of HER2-driven breast cancers. However, ~30% of patients develop resistance to trastuzumab and recur; and nearly all patients with advanced disease develop resistance over time and succumb to the disease. Mechanisms of trastuzumab resistance (TzR) are not well understood, although some studies suggest that growth factor signaling through other receptors may be responsible. However, these studies were based on cell culture models of the disease, and thus, it is not known which pathways are driving the resistance in vivo. Using an integrative transcriptomic approach of RNA isolated from trastuzumab-sensitive and trastuzumab-resistant HER2+ tumors, and isogenic cell culture models, we identified a small set of mRNAs and lincRNAs that are associated with trastuzumab-resistance (TzR). Functional analysis of a top candidate gene, S100P, demonstrated that inhibition of S100P results in reversing TzR. Mechanistically, S100P activates the RAS/MEK/MAPK pathway to compensate for HER2 inhibition by trastuzumab. Finally, we demonstrated that the upregulation of S100P appears to be driven by epigenomic changes at the enhancer level. Our current findings should pave the path toward new therapies for breast cancer patients.


Subject(s)
Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Receptor, ErbB-2/genetics , Transcriptome , Trastuzumab/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA Interference , Receptor, ErbB-2/metabolism , Signal Transduction/genetics , Trastuzumab/pharmacology
12.
Cell Stem Cell ; 14(6): 854-63, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24905169

ABSTRACT

Naive mouse embryonic stem cells (mESCs) and primed epiblast stem cells (mEpiSCs) represent successive snapshots of pluripotency during embryogenesis. Using transcriptomic and epigenomic mapping we show that a small fraction of transcripts are differentially expressed between mESCs and mEpiSCs and that these genes show expected changes in chromatin at their promoters and enhancers. Unexpectedly, the cis-regulatory circuitry of genes that are expressed at identical levels between these cell states also differs dramatically. In mESCs, these genes are associated with dominant proximal enhancers and dormant distal enhancers, which we term seed enhancers. In mEpiSCs, the naive-dominant enhancers are lost, and the seed enhancers take up primary transcriptional control. Seed enhancers have increased sequence conservation and show preferential usage in downstream somatic tissues, often expanding into super enhancers. We propose that seed enhancers ensure proper enhancer utilization and transcriptional fidelity as mammalian cells transition from naive pluripotency to a somatic regulatory program.


Subject(s)
Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Mice
13.
Development ; 141(5): 1129-39, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24550119

ABSTRACT

Trithorax (TRX) antagonizes epigenetic silencing by Polycomb group (PcG) proteins, stimulates enhancer-dependent transcription, and establishes a 'cellular memory' of active transcription of PcG-regulated genes. The mechanisms underlying these TRX functions remain largely unknown, but are presumed to involve its histone H3K4 methyltransferase activity. We report that the SET domains of TRX and TRX-related (TRR) have robust histone H3K4 monomethyltransferase activity in vitro and that Tyr3701 of TRX and Tyr2404 of TRR prevent them from being trimethyltransferases. The trx(Z11) missense mutation (G3601S), which abolishes H3K4 methyltransferase activity in vitro, reduces the H3K4me1 but not the H3K4me3 level in vivo. trx(Z11) also suppresses the impaired silencing phenotypes of the Pc(3) mutant, suggesting that H3K4me1 is involved in antagonizing Polycomb silencing. Polycomb silencing is also antagonized by TRX-dependent H3K27 acetylation by CREB-binding protein (CBP). We show that perturbation of Polycomb silencing by TRX overexpression requires CBP. We also show that TRX and TRR are each physically associated with CBP in vivo, that TRX binds directly to the CBP KIX domain, and that the chromatin binding patterns of TRX and TRR are highly correlated with CBP and H3K4me1 genome-wide. In vitro acetylation of H3K27 by CBP is enhanced on K4me1-containing H3 substrates, and independently altering the H3K4me1 level in vivo, via the H3K4 demethylase LSD1, produces concordant changes in H3K27ac. These data indicate that the catalytic activities of TRX and CBP are physically coupled and suggest that both activities play roles in antagonizing Polycomb silencing, stimulating enhancer activity and cellular memory.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Histones/metabolism , Polycomb Repressive Complex 1/metabolism , Acetylation , Animals , Chromosomal Proteins, Non-Histone/genetics , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Gene Silencing/physiology , Methylation , Polycomb Repressive Complex 1/genetics , Protein Binding/genetics , Protein Binding/physiology
14.
Genome Res ; 24(1): 1-13, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24196873

ABSTRACT

DNA variants (SNPs) that predispose to common traits often localize within noncoding regulatory elements such as enhancers. Moreover, loci identified by genome-wide association studies (GWAS) often contain multiple SNPs in linkage disequilibrium (LD), any of which may be causal. Thus, determining the effect of these multiple variant SNPs on target transcript levels has been a major challenge. Here, we provide evidence that for six common autoimmune disorders (rheumatoid arthritis, Crohn's disease, celiac disease, multiple sclerosis, lupus, and ulcerative colitis), the GWAS association arises from multiple polymorphisms in LD that map to clusters of enhancer elements active in the same cell type. This finding suggests a "multiple enhancer variant" hypothesis for common traits, where several variants in LD impact multiple enhancers and cooperatively affect gene expression. Using a novel method to delineate enhancer-gene interactions, we show that multiple enhancer variants within a given locus typically target the same gene. Using available data from HapMap and B lymphoblasts as a model system, we provide evidence at numerous loci that multiple enhancer variants cooperatively contribute to altered expression of their gene targets. The effects on target transcript levels tend to be modest and can be either gain- or loss-of-function. Additionally, the genes associated with multiple enhancer variants encode proteins that are often functionally related and enriched in common pathways. Overall, the multiple enhancer variant hypothesis offers a new paradigm by which noncoding variants can confer susceptibility to common traits.


Subject(s)
Autoimmune Diseases/genetics , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Linkage Disequilibrium , Arthritis, Rheumatoid/genetics , Celiac Disease/genetics , Colitis, Ulcerative/genetics , Crohn Disease/genetics , Gene Expression , Genetic Variation , Genome-Wide Association Study , Humans , Lupus Erythematosus, Systemic/genetics , Multiple Sclerosis/genetics , Phenotype , Polymorphism, Single Nucleotide , Quantitative Trait Loci
15.
Science ; 336(6082): 736-9, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22499810

ABSTRACT

Cancer is characterized by gene expression aberrations. Studies have largely focused on coding sequences and promoters, even though distal regulatory elements play a central role in controlling transcription patterns. We used the histone mark H3K4me1 to analyze gain and loss of enhancer activity genome-wide in primary colon cancer lines relative to normal colon crypts. We identified thousands of variant enhancer loci (VELs) that comprise a signature that is robustly predictive of the in vivo colon cancer transcriptome. Furthermore, VELs are enriched in haplotype blocks containing colon cancer genetic risk variants, implicating these genomic regions in colon cancer pathogenesis. We propose that reproducible changes in the epigenome at enhancer elements drive a specific transcriptional program to promote colon carcinogenesis.


Subject(s)
Colonic Neoplasms/genetics , Enhancer Elements, Genetic , Epigenesis, Genetic , Histones/metabolism , Transcriptome , Cell Line, Tumor , Chromatin Immunoprecipitation , Colon/metabolism , Colonic Neoplasms/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Genetic Loci , Humans , Intestinal Mucosa/metabolism , Methylation , Polymorphism, Single Nucleotide , Promoter Regions, Genetic
16.
Nucleic Acids Res ; 39(12): 4949-60, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21355038

ABSTRACT

The transcription of ribosomal RNA (rRNA) is critical to life. Despite its importance, ribosomal DNA (rDNA) is not included in current genome assemblies and, consequently, genomic analyses to date have excluded rDNA. Here, we show that short sequence reads can be aligned to a genome assembly containing a single rDNA repeat. Integrated analysis of ChIP-seq, DNase-seq, MNase-seq and RNA-seq data reveals several novel findings. First, the coding region of active rDNA is contained within nucleosome-depleted open chromatin that is highly transcriptionally active. Second, histone modifications are located not only at the rDNA promoter but also at novel sites within the intergenic spacer. Third, the distributions of active modifications are more similar within and between different cell types than repressive modifications. Fourth, UBF, a positive regulator of rRNA transcription, binds to sites throughout the genome. Lastly, the insulator binding protein CTCF associates with the spacer promoter of rDNA, suggesting that transcriptional insulation plays a role in regulating the transcription of rRNA. Taken together, these analyses confirm and expand the results of previous ChIP studies of rDNA and provide novel avenues for exploration of chromatin-mediated regulation of rDNA.


Subject(s)
DNA, Ribosomal/chemistry , Genome, Human , RNA, Ribosomal/biosynthesis , CCCTC-Binding Factor , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA, Ribosomal/metabolism , Genomics , HEK293 Cells , High-Throughput Nucleotide Sequencing , Histones/metabolism , Humans , K562 Cells , Nucleosomes/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism , Promoter Regions, Genetic , RNA Polymerase I/metabolism , Repressor Proteins/metabolism , Sequence Alignment , Sequence Analysis, DNA , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...