Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Mol Cell ; 82(16): 3030-3044.e8, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35764091

ABSTRACT

Characterized by intracellular lipid droplet accumulation, clear cell renal cell carcinoma (ccRCC) is resistant to cytotoxic chemotherapy and is a lethal disease. Through an unbiased siRNA screen of 2-oxoglutarate (2-OG)-dependent enzymes, which play a critical role in tumorigenesis, we identified Jumonji domain-containing 6 (JMJD6) as an essential gene for ccRCC tumor development. The downregulation of JMJD6 abolished ccRCC colony formation in vitro and inhibited orthotopic tumor growth in vivo. Integrated ChIP-seq and RNA-seq analyses uncovered diacylglycerol O-acyltransferase 1 (DGAT1) as a critical JMJD6 effector. Mechanistically, JMJD6 interacted with RBM39 and co-occupied DGAT1 gene promoter with H3K4me3 to induce DGAT1 expression. JMJD6 silencing reduced DGAT1, leading to decreased lipid droplet formation and tumorigenesis. The pharmacological inhibition (or depletion) of DGAT1 inhibited lipid droplet formation in vitro and ccRCC tumorigenesis in vivo. Thus, the JMJD6-DGAT1 axis represents a potential new therapeutic target for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Diacylglycerol O-Acyltransferase , Jumonji Domain-Containing Histone Demethylases , Kidney Neoplasms , Carcinogenesis/genetics , Carcinoma, Renal Cell/genetics , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Epigenesis, Genetic , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Kidney Neoplasms/genetics , Lipid Droplets/metabolism
2.
Mol Cell ; 77(6): 1294-1306.e5, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32023483

ABSTRACT

von Hippel-Lindau (VHL) is a critical tumor suppressor in clear cell renal cell carcinomas (ccRCCs). It is important to identify additional therapeutic targets in ccRCC downstream of VHL loss besides hypoxia-inducible factor 2α (HIF2α). By performing a genome-wide screen, we identified Scm-like with four malignant brain tumor domains 1 (SFMBT1) as a candidate pVHL target. SFMBT1 was considered to be a transcriptional repressor but its role in cancer remains unclear. ccRCC patients with VHL loss-of-function mutations displayed elevated SFMBT1 protein levels. SFMBT1 hydroxylation on Proline residue 651 by EglN1 mediated its ubiquitination and degradation governed by pVHL. Depletion of SFMBT1 abolished ccRCC cell proliferation in vitro and inhibited orthotopic tumor growth in vivo. Integrated analyses of ChIP-seq, RNA-seq, and patient prognosis identified sphingosine kinase 1 (SPHK1) as a key SFMBT1 target gene contributing to its oncogenic phenotype. Therefore, the pVHL-SFMBT1-SPHK1 axis serves as a potential therapeutic avenue for ccRCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Repressor Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Genome-Wide Association Study , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prognosis , Prolyl Hydroxylases/genetics , Prolyl Hydroxylases/metabolism , Repressor Proteins/genetics , Tumor Cells, Cultured , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Xenograft Model Antitumor Assays
3.
Hum Mol Genet ; 33(5): 448-464, 2024 Feb 18.
Article in English | MEDLINE | ID: mdl-37975905

ABSTRACT

Biallelic mutations in interphotoreceptor matrix proteoglycan 2 (IMPG2) in humans cause retinitis pigmentosa (RP) with early macular involvement, albeit the disease progression varies widely due to genetic heterogeneity and IMPG2 mutation type. There are currently no treatments for IMPG2-RP. To aid preclinical studies toward eventual treatments, there is a need to better understand the progression of disease pathology in appropriate animal models. Toward this goal, we developed mouse models with patient mimicking homozygous frameshift (T807Ter) or missense (Y250C) Impg2 mutations, as well as mice with a homozygous frameshift mutation (Q244Ter) designed to completely prevent IMPG2 protein expression, and characterized the trajectory of their retinal pathologies across postnatal development until late adulthood. We found that the Impg2T807Ter/T807Ter and Impg2Q244Ter/Q244Ter mice exhibited early onset gliosis, impaired photoreceptor outer segment maintenance, appearance of subretinal deposits near the optic disc, disruption of the outer retina, and neurosensorial detachment, whereas the Impg2Y250C/Y250C mice exhibited minimal retinal pathology. These results demonstrate the importance of mutation type in disease progression in IMPG2-RP and provide a toolkit and preclinical data for advancing therapeutic approaches.


Subject(s)
Proteoglycans , Retinitis Pigmentosa , Humans , Animals , Mice , Adult , Proteoglycans/genetics , Retina , Mutation , Retinitis Pigmentosa/genetics , Disease Progression
4.
Nature ; 587(7833): 281-284, 2020 11.
Article in English | MEDLINE | ID: mdl-33087932

ABSTRACT

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by a mutation or deletion of the maternally inherited UBE3A allele. In neurons, the paternally inherited UBE3A allele is silenced in cis by a long non-coding RNA called UBE3A-ATS. Here, as part of a systematic screen, we found that Cas9 can be used to activate ('unsilence') paternal Ube3a in cultured mouse and human neurons when targeted to Snord115 genes, which are small nucleolar RNAs that are clustered in the 3' region of Ube3a-ATS. A short Cas9 variant and guide RNA that target about 75 Snord115 genes were packaged into an adeno-associated virus and administered to a mouse model of AS during the embryonic and early postnatal stages, when the therapeutic benefit of restoring Ube3a is predicted to be greatest1,2. This early treatment unsilenced paternal Ube3a throughout the brain for at least 17 months and rescued anatomical and behavioural phenotypes in AS mice. Genomic integration of the adeno-associated virus vector into Cas9 target sites caused premature termination of Ube3a-ATS at the vector-derived polyA cassette, or when integrated in the reverse orientation, by transcriptional collision with the vector-derived Cas9 transcript. Our study shows that targeted genomic integration of a gene therapy vector can restore the function of paternally inherited UBE3A throughout life, providing a path towards a disease-modifying treatment for a syndromic neurodevelopmental disorder.


Subject(s)
Angelman Syndrome/genetics , Angelman Syndrome/therapy , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Gene Editing , Genetic Therapy/methods , RNA, Long Noncoding/genetics , Ubiquitin-Protein Ligases/genetics , Animals , CRISPR-Associated Protein 9/genetics , Dependovirus/genetics , Disease Models, Animal , Female , Gene Silencing , Genetic Vectors/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Nervous System/metabolism , Paternal Inheritance/genetics , Phenotype , RNA, Guide, Kinetoplastida/genetics
5.
Proc Natl Acad Sci U S A ; 119(36): e2119854119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037364

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is characterized by the loss of tumor suppressor Von Hippel Lindau (VHL) function. VHL is the component of an E3 ligase complex that promotes the ubiquitination and degradation of hypoxia inducible factor α (HIF-α) (including HIF1α and HIF2α) and Zinc Fingers And Homeoboxes 2 (ZHX2). Our recent research showed that ZHX2 contributed to ccRCC tumorigenesis in a HIF-independent manner. However, it is still unknown whether ZHX2 could be modified through deubiquitination even in the absence of pVHL. Here, we performed a deubiquitinase (DUB) complementary DNA (cDNA) library binding screen and identified USP13 as a DUB that bound ZHX2 and promoted ZHX2 deubiquitination. As a result, USP13 promoted ZHX2 protein stability in an enzymatically dependent manner, and depletion of USP13 led to ZHX2 down-regulation in ccRCC. Functionally, USP13 depletion led to decreased cell proliferation measured by two-dimensional (2D) colony formation and three-dimensional (3D) anchorage-independent growth. Furthermore, USP13 was essential for ccRCC tumor growth in vivo, and the effect was partially mediated by its regulation on ZHX2. Our findings support that USP13 may be a key effector in ccRCC tumorigenesis.


Subject(s)
Carcinoma, Renal Cell , Homeodomain Proteins , Kidney Neoplasms , Transcription Factors , Ubiquitin-Specific Proteases , Carcinogenesis/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
6.
Proc Natl Acad Sci U S A ; 117(23): 13023-13032, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32461361

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is characterized by loss of tumor suppressor Von Hippel Lindau (VHL) function, which leads to accumulation of hypoxia inducible factor α (including HIF1α and HIF2α). HIF2α was previously reported to be one of the major oncogenic drivers in ccRCC, however, its therapeutic targets remain challenging. Here we performed a deubiquitinase (DUB) complementary DNA (cDNA) library binding screen and discovered that ubiquitin-specific peptidase 37 (USP37) is a DUB that binds HIF2α and promotes HIF2α deubiquitination. As a result, USP37 promotes HIF2α protein stability in an enzymatically dependent manner, and depletion of USP37 leads to HIF2α down-regulation in ccRCC. Functionally, USP37 depletion causes decreased cell proliferation measured by MTS, two-dimensional (2D) colony formation as well as three-dimensional (3D) anchorage- independent growth. USP37 is also essential for maintaining kidney tumorigenesis in an orthotopic xenograft model and its depletion leads to both decreased primary kidney tumorigenesis and spontaneous lung metastasis. Our results suggest that USP37 is a potential therapeutic target in ccRCC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/pathology , Endopeptidases/metabolism , Kidney Neoplasms/pathology , Animals , Carcinogenesis , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation Sequencing , Down-Regulation , Endopeptidases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Mice , Protein Stability , RNA, Small Interfering/metabolism , RNA-Seq , Ubiquitination , Xenograft Model Antitumor Assays
7.
PLoS Genet ; 16(1): e1008537, 2020 01.
Article in English | MEDLINE | ID: mdl-31961859

ABSTRACT

Gene transcription profiles across tissues are largely defined by the activity of regulatory elements, most of which correspond to regions of accessible chromatin. Regulatory element activity is in turn modulated by genetic variation, resulting in variable transcription rates across individuals. The interplay of these factors, however, is poorly understood. Here we characterize expression and chromatin state dynamics across three tissues-liver, lung, and kidney-in 47 strains of the Collaborative Cross (CC) mouse population, examining the regulation of these dynamics by expression quantitative trait loci (eQTL) and chromatin QTL (cQTL). QTL whose allelic effects were consistent across tissues were detected for 1,101 genes and 133 chromatin regions. Also detected were eQTL and cQTL whose allelic effects differed across tissues, including local-eQTL for Pik3c2g detected in all three tissues but with distinct allelic effects. Leveraging overlapping measurements of gene expression and chromatin accessibility on the same mice from multiple tissues, we used mediation analysis to identify chromatin and gene expression intermediates of eQTL effects. Based on QTL and mediation analyses over multiple tissues, we propose a causal model for the distal genetic regulation of Akr1e1, a gene involved in glycogen metabolism, through the zinc finger transcription factor Zfp985 and chromatin intermediates. This analysis demonstrates the complexity of transcriptional and chromatin dynamics and their regulation over multiple tissues, as well as the value of the CC and related genetic resource populations for identifying specific regulatory mechanisms within cells and tissues.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/chemistry , Quantitative Trait Loci , Animals , Chromatin/genetics , Chromatin/metabolism , Kidney/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Liver/metabolism , Lung/metabolism , Male , Mice , Organ Specificity , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism
8.
J Pathol ; 252(2): 125-137, 2020 10.
Article in English | MEDLINE | ID: mdl-32619021

ABSTRACT

Activation of the nuclear factor (erythroid-derived 2)-like 2 (NFE2L2 or NRF2) transcription factor is a critical and evolutionarily conserved cellular response to oxidative stress, metabolic stress, and xenobiotic insult. Deficiency of NRF2 results in hypersensitivity to a variety of stressors, whereas its aberrant activation contributes to several cancer types, most commonly squamous cell carcinomas of the esophagus, oral cavity, bladder, and lung. Between 10% and 35% of patients with squamous cell carcinomas display hyperactive NRF2 signaling, harboring activating mutations and copy number amplifications of the NFE2L2 oncogene or inactivating mutations or deletions of KEAP1 or CUL3, the proteins of which co-complex to ubiquitylate and degrade NRF2 protein. To better understand the role of NRF2 in tumorigenesis and more broadly in development, we engineered the endogenous Nfe2l2 genomic locus to create a conditional mutant LSL-Nrf2E79Q mouse model. The E79Q mutation, one of the most commonly observed NRF2-activating mutations in human squamous cancers, codes for a mutant protein that does not undergo KEAP1/CUL3-dependent degradation, resulting in its constitutive activity. Expression of NRF2 E79Q protein in keratin 14 (KRT14)-positive murine tissues resulted in hyperplasia of squamous cell tissues of the tongue, forestomach, and esophagus, a stunted body axis, decreased weight, and decreased visceral adipose depots. RNA-seq profiling and follow-up validation studies of cultured NRF2E79Q murine esophageal epithelial cells revealed known and novel NRF2-regulated transcriptional programs, including genes associated with squamous cell carcinoma (e.g. Myc), lipid and cellular metabolism (Hk2, Ppard), and growth factors (Areg, Bmp6, Vegfa). These data suggest that in addition to decreasing adipogenesis, KRT14-restricted NRF2 activation drives hyperplasia of the esophagus, forestomach, and tongue, but not formation of squamous cell carcinoma. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Adipose Tissue, White/pathology , Carcinogenesis/genetics , Disease Models, Animal , NF-E2-Related Factor 2/genetics , Precancerous Conditions/genetics , Upper Gastrointestinal Tract/pathology , Animals , Carcinoma, Squamous Cell/genetics , Esophagus/pathology , Humans , Hyperplasia/genetics , Mice , Mutation , Tongue/pathology
9.
Alcohol Clin Exp Res ; 44(8): 1540-1550, 2020 08.
Article in English | MEDLINE | ID: mdl-32557641

ABSTRACT

BACKGROUND: Early gestational alcohol exposure is associated with severe craniofacial and CNS dysmorphologies and behavioral abnormalities during adolescence and adulthood. Alcohol exposure during the formation of the neural tube (gestational day [GD] 8 to 10 in mice; equivalent to4th week of human pregnancy) disrupts development of ventral midline brain structures such as the pituitary, septum, and ventricles. This study identifies transcriptomic changes in the rostroventral neural tube (RVNT), the region of the neural tube that gives rise to the midline structures sensitive to alcohol exposure during neurulation. METHODS: Female C57BL/6J mice were administered 2 doses of alcohol (2.9 g/kg) or vehicle 4 hours apart on GD 9.0. The RVNTs of embryos were collected 6 or 24 hours after the first dose and processed for RNA-seq. RESULTS: Six hours following GD 9.0 alcohol exposure (GD 9.25), over 2,300 genes in the RVNT were determined to be differentially regulated by alcohol. Enrichment analysis determined that PAE affected pathways related to cell proliferation, p53 signaling, ribosome biogenesis, and immune activation. In addition, over 100 genes involved in primary cilia formation and function and regulation of morphogenic pathways were altered 6 hours after alcohol exposure. The changes to gene expression were largely transient, as only 91 genes identified as differentially regulated by prenatal alcohol at GD 10 (24 hours postexposure). Functionally, the differentially regulated genes at GD 10 were related to organogenesis and cell migration. CONCLUSIONS: These data give a comprehensive view of the changing landscape of the embryonic transcriptome networks in regions of the neural tube that give rise to brain structures impacted by a neurulation-stage alcohol exposure. Identification of gene networks dysregulated by alcohol will help elucidate the pathogenic mechanisms of alcohol's actions.


Subject(s)
Central Nervous System Depressants/pharmacology , Embryo, Mammalian/drug effects , Ethanol/pharmacology , Gene Expression Regulation, Developmental/drug effects , Neural Tube/drug effects , Neurulation/drug effects , Animals , Cell Proliferation/genetics , Cilia/genetics , Embryo, Mammalian/metabolism , Female , Gene Expression Profiling , Mice , Neural Tube/metabolism , Neurulation/genetics , Organelle Biogenesis , Pregnancy , RNA-Seq , Ribosomes/genetics , Tumor Suppressor Protein p53
10.
Proc Natl Acad Sci U S A ; 113(11): 3018-23, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929321

ABSTRACT

Mutations in chromatin-modifying proteins and transcription factors are commonly associated with a wide variety of cancers. Through gain- or loss-of-function, these mutations may result in characteristic alterations of accessible chromatin, indicative of shifts in the landscape of regulatory elements genome-wide. The identification of compounds that reverse a specific chromatin signature could lead to chemical probes or potential therapies. To explore whether chromatin accessibility could serve as a platform for small molecule screening, we adapted formaldehyde-assisted isolation of regulatory elements (FAIRE), a chemical method to enrich for nucleosome-depleted genomic regions, as a high-throughput, automated assay. After demonstrating the validity and robustness of this approach, we applied this method to screen an epigenetically targeted small molecule library by evaluating regions of aberrant nucleosome depletion mediated by EWSR1-FLI1, the chimeric transcription factor critical for the bone and soft tissue tumor Ewing sarcoma. As a class, histone deacetylase inhibitors were greatly overrepresented among active compounds. These compounds resulted in diminished accessibility at targeted sites by disrupting transcription of EWSR1-FLI1. Capitalizing on precise differences in chromatin accessibility for drug discovery efforts offers significant advantages because it does not depend on the a priori selection of a single molecular target and may detect novel biologically relevant pathways.


Subject(s)
Chromatin/drug effects , High-Throughput Screening Assays/methods , Oncogene Proteins, Fusion/antagonists & inhibitors , Transcription, Genetic/drug effects , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Bone Neoplasms/pathology , Cell Line, Tumor , Chromatin/ultrastructure , Drug Design , Drug Evaluation, Preclinical , Histone Deacetylase Inhibitors/isolation & purification , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Molecular Targeted Therapy , Nucleosomes/ultrastructure , Oncogene Proteins, Fusion/genetics , Panobinostat , Phenylbutyrates/pharmacology , Sarcoma, Ewing/pathology , Small Molecule Libraries , Vorinostat
11.
Gut ; 67(1): 36-42, 2018 01.
Article in English | MEDLINE | ID: mdl-27742763

ABSTRACT

OBJECTIVE: The clinical presentation and course of Crohn's disease (CD) is highly variable. We sought to better understand the cellular and molecular mechanisms that guide this heterogeneity, and characterise the cellular processes associated with disease phenotypes. DESIGN: We examined both gene expression and gene regulation (chromatin accessibility) in non-inflamed colon tissue from a cohort of adult patients with CD and control patients. To support the generality of our findings, we analysed previously published expression data from a large cohort of treatment-naïve paediatric CD and control ileum. RESULTS: We found that adult patients with CD clearly segregated into two classes based on colon tissue gene expression-one that largely resembled the normal colon and one where certain genes showed expression patterns normally specific to the ileum. These classes were supported by changes in gene regulatory profiles observed at the level of chromatin accessibility, reflective of a fundamental shift in underlying molecular phenotypes. Furthermore, gene expression from the ilea of a treatment-naïve cohort of paediatric patients with CD could be similarly subdivided into colon-like and ileum-like classes. Finally, expression patterns within these CD subclasses highlight large-scale differences in the immune response and aspects of cellular metabolism, and were associated with multiple clinical phenotypes describing disease behaviour, including rectal disease and need for colectomy. CONCLUSIONS: Our results strongly suggest that these molecular signatures define two clinically relevant forms of CD irrespective of tissue sampling location, patient age or treatment status.


Subject(s)
Crohn Disease/genetics , Adult , Age Factors , Case-Control Studies , Child , Colon/metabolism , Crohn Disease/classification , Crohn Disease/metabolism , Crohn Disease/therapy , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Genome-Wide Association Study , Humans , Ileum/metabolism , Male , Phenotype , Principal Component Analysis , Prognosis
12.
J Neurosci ; 37(42): 10230-10239, 2017 10 18.
Article in English | MEDLINE | ID: mdl-28931574

ABSTRACT

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by mutation or deletion of the maternal UBE3A allele. The maternal UBE3A allele is expressed in nearly all neurons of the brain and spinal cord, whereas the paternal UBE3A allele is repressed by an extremely long antisense transcript (UBE3A-ATS). Little is known about expression of UBE3A in the peripheral nervous system, where loss of maternal UBE3A might contribute to AS phenotypes. Here we sought to examine maternal and paternal Ube3a expression in DRGs neurons and to evaluate whether nociceptive responses were affected in AS model mice (global deletion of maternal Ube3a allele; Ube3am-/p+). We found that most large-diameter proprioceptive and mechanosensitive DRG neurons expressed maternal Ube3a and paternal Ube3a-ATS In contrast, most small-diameter neurons expressed Ube3a biallelically and had low to undetectable levels of Ube3a-ATS Analysis of single-cell DRG transcriptomes further suggested that Ube3a is expressed monoallelically in myelinated large-diameter neurons and biallelically in unmyelinated small-diameter neurons. Behavioral responses to some noxious thermal and mechanical stimuli were enhanced in male and female AS model mice; however, nociceptive responses were not altered by the conditional deletion of maternal Ube3a in the DRG. These data suggest that the enhanced nociceptive responses in AS model mice are due to loss of maternal Ube3a in the central, but not peripheral, nervous system. Our study provides new insights into sensory processing deficits associated with AS.SIGNIFICANCE STATEMENT Angelman syndrome (AS) is a neurodevelopmental disorder caused by loss or mutation of the maternal UBE3A allele. While sensory processing deficits are frequently associated with AS, it is currently unknown whether Ube3a is expressed in peripheral sensory neurons or whether maternal deletion of Ube3a affects somatosensory responses. Here, we found that Ube3a is primarily expressed from the maternally inherited allele in myelinated large-diameter sensory neurons and biallelically expressed in unmyelinated small-diameter neurons. Nociceptive responses to select noxious thermal and mechanical stimuli were enhanced following global, but not sensory neuron-specific, deletion of maternal Ube3a in mice. These data suggest that maternal loss of Ube3a affects nociception via a central, but not peripheral mechanism, with implications for AS.


Subject(s)
Angelman Syndrome/genetics , Angelman Syndrome/pathology , Disease Models, Animal , Pain Measurement/methods , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Animals , Female , Ganglia, Spinal/pathology , Ganglia, Spinal/physiology , Male , Mice , Mice, Knockout , Spinal Cord/pathology , Spinal Cord/physiology
13.
Mamm Genome ; 29(1-2): 153-167, 2018 02.
Article in English | MEDLINE | ID: mdl-29429127

ABSTRACT

Epigenetic effects of environmental chemicals are under intense investigation to fill existing knowledge gaps between environmental/occupational exposures and adverse health outcomes. Chromatin accessibility is one prominent mechanism of epigenetic control of transcription, and understanding of the chemical effects on both could inform the causal role of epigenetic alterations in disease mechanisms. In this study, we hypothesized that baseline variability in chromatin organization and transcription profiles among various tissues and mouse strains influence the outcome of exposure to the DNA damaging chemical 1,3-butadiene. To test this hypothesis, we evaluated DNA damage along with comprehensive quantification of RNA transcripts (RNA-seq), identification of accessible chromatin (ATAC-seq), and characterization of regions with histone modifications associated with active transcription (ChIP-seq for acetylation at histone 3 lysine 27, H3K27ac). We collected these data in the lung, liver, and kidney of mice from two genetically divergent strains, C57BL/6J and CAST/EiJ, that were exposed to clean air or to 1,3-butadiene (~600 ppm) for 2 weeks. We found that tissue effects dominate differences in both gene expression and chromatin states, followed by strain effects. At baseline, xenobiotic metabolism was consistently more active in CAST/EiJ, while immune system pathways were more active in C57BL/6J across tissues. Surprisingly, even though all three tissues in both strains harbored butadiene-induced DNA damage, little transcriptional effect of butadiene was observed in liver and kidney. Toxicologically relevant effects of butadiene in the lung were on the pathways of xenobiotic metabolism and inflammation. We also found that variability in chromatin accessibility across individuals (i.e., strains) only partially explains the variability in transcription. This study showed that variation in the basal states of epigenome and transcriptome may be useful indicators for individuals or tissues susceptible to genotoxic environmental chemicals.


Subject(s)
DNA Damage/drug effects , Epigenesis, Genetic , Transcription, Genetic/genetics , Transcriptome/genetics , Animals , Butadienes/toxicity , Carcinogens/toxicity , Chromatin/drug effects , Histones/genetics , Liver/drug effects , Liver/pathology , Lung/drug effects , Lung/pathology , Mice , Mutagenicity Tests , Organ Specificity/drug effects , Transcription, Genetic/drug effects
14.
Nature ; 489(7414): 75-82, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22955617

ABSTRACT

DNase I hypersensitive sites (DHSs) are markers of regulatory DNA and have underpinned the discovery of all classes of cis-regulatory elements including enhancers, promoters, insulators, silencers and locus control regions. Here we present the first extensive map of human DHSs identified through genome-wide profiling in 125 diverse cell and tissue types. We identify ∼2.9 million DHSs that encompass virtually all known experimentally validated cis-regulatory sequences and expose a vast trove of novel elements, most with highly cell-selective regulation. Annotating these elements using ENCODE data reveals novel relationships between chromatin accessibility, transcription, DNA methylation and regulatory factor occupancy patterns. We connect ∼580,000 distal DHSs with their target promoters, revealing systematic pairing of different classes of distal DHSs and specific promoter types. Patterning of chromatin accessibility at many regulatory regions is organized with dozens to hundreds of co-activated elements, and the transcellular DNase I sensitivity pattern at a given region can predict cell-type-specific functional behaviours. The DHS landscape shows signatures of recent functional evolutionary constraint. However, the DHS compartment in pluripotent and immortalized cells exhibits higher mutation rates than that in highly differentiated cells, exposing an unexpected link between chromatin accessibility, proliferative potential and patterns of human variation.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , DNA/genetics , Encyclopedias as Topic , Genome, Human/genetics , Molecular Sequence Annotation , Regulatory Sequences, Nucleic Acid/genetics , DNA Footprinting , DNA Methylation , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/metabolism , Evolution, Molecular , Genomics , Humans , Mutation Rate , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Transcription Initiation Site , Transcription, Genetic
15.
Eur J Immunol ; 46(8): 1912-25, 2016 08.
Article in English | MEDLINE | ID: mdl-27159132

ABSTRACT

Intestinal macrophages (IMs) are uniquely programmed to tolerate exposure to bacteria without mounting potent inflammatory responses. The cytokine IL-10 maintains the macrophage anti-inflammatory response such that loss of IL-10 results in chronic intestinal inflammation. To investigate how IL-10-deficiency alters IM programming and bacterial tolerance, we studied changes in chromatin accessibility in response to bacteria in macrophages from two distinct niches, the intestine and bone-marrow, from both wild-type and IL-10-deficient (Il10(-/-) ) mice. We identified chromatin accessibility changes associated with bacterial exposure and IL-10 deficiency in both bone marrow derived macrophages and IMs. Surprisingly, Il10(-/-) IMs adopted chromatin and gene expression patterns characteristic of an inflammatory response, even in the absence of bacteria. Further, when recombinant IL-10 was added to Il10(-/-) cells, it could not revert the chromatin landscape to a normal state. Our results demonstrate that IL-10 deficiency results in stable chromatin alterations in macrophages, even in the absence of bacteria. This supports a model in which IL-10-deficiency leads to chromatin alterations that contribute to a loss of IM tolerance to bacteria, which is a primary initiating event in chronic intestinal inflammation.


Subject(s)
Chromatin/metabolism , Inflammation/immunology , Interleukin-10/genetics , Intestines/physiopathology , Macrophages/metabolism , Animals , Cytokines/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression , Humans , Immune Tolerance , Intestines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Genome Res ; 24(2): 241-50, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24158655

ABSTRACT

Comprehensive sequencing of human cancers has identified recurrent mutations in genes encoding chromatin regulatory proteins. For clear cell renal cell carcinoma (ccRCC), three of the five commonly mutated genes encode the chromatin regulators PBRM1, SETD2, and BAP1. How these mutations alter the chromatin landscape and transcriptional program in ccRCC or other cancers is not understood. Here, we identified alterations in chromatin organization and transcript profiles associated with mutations in chromatin regulators in a large cohort of primary human kidney tumors. By associating variation in chromatin organization with mutations in SETD2, which encodes the enzyme responsible for H3K36 trimethylation, we found that changes in chromatin accessibility occurred primarily within actively transcribed genes. This increase in chromatin accessibility was linked with widespread alterations in RNA processing, including intron retention and aberrant splicing, affecting ∼25% of all expressed genes. Furthermore, decreased nucleosome occupancy proximal to misspliced exons was observed in tumors lacking H3K36me3. These results directly link mutations in SETD2 to chromatin accessibility changes and RNA processing defects in cancer. Detecting the functional consequences of specific mutations in chromatin regulatory proteins in primary human samples could ultimately inform the therapeutic application of an emerging class of chromatin-targeted compounds.


Subject(s)
Carcinoma, Renal Cell/genetics , Chromatin/genetics , Histone-Lysine N-Methyltransferase/genetics , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/pathology , DNA-Binding Proteins , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/metabolism , Humans , Kidney Neoplasms/pathology , Mutation , Nuclear Proteins/genetics , RNA Processing, Post-Transcriptional/genetics , RNA Splicing/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics
17.
J Biol Chem ; 290(43): 26088-102, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26338712

ABSTRACT

G9a and GLP lysine methyltransferases form a heterodimeric complex that is responsible for the majority of histone H3 lysine 9 mono- and di-methylation (H3K9me1/me2). Widely interspaced zinc finger (WIZ) associates with the G9a-GLP protein complex, but its role in mediating lysine methylation is poorly defined. Here, we show that WIZ regulates global H3K9me2 levels by facilitating the interaction of G9a with chromatin. Disrupting the association of G9a-GLP with chromatin by depleting WIZ resulted in altered gene expression and protein-protein interactions that were distinguishable from that of small molecule-based inhibition of G9a/GLP, supporting discrete functions of the G9a-GLP-WIZ chromatin complex in addition to H3K9me2 methylation.


Subject(s)
Chromatin/metabolism , Kruppel-Like Transcription Factors/physiology , Methyltransferases/metabolism , Chromatin Immunoprecipitation , DNA Methylation , Gene Expression/genetics , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Kruppel-Like Transcription Factors/genetics , Protein Binding
18.
Genome Res ; 22(2): 259-70, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22086061

ABSTRACT

Chromosomal translocations involving transcription factor genes have been identified in an increasingly wide range of cancers. Some translocations can create a protein "chimera" that is composed of parts from different proteins. How such chimeras cause cancer, and why they cause cancer in some cell types but not others, is not understood. One such chimera is EWS-FLI, the most frequently occurring translocation in Ewing Sarcoma, a malignant bone and soft tissue tumor of children and young adults. Using EWS-FLI and its parental transcription factor, FLI1, we created a unique experimental system to address questions regarding the genomic mechanisms by which chimeric transcription factors cause cancer. We found that in tumor cells, EWS-FLI targets regions of the genome distinct from FLI1, despite identical DNA-binding domains. In primary endothelial cells, however, EWS-FLI and FLI1 demonstrate similar targeting. To understand this mistargeting, we examined chromatin organization. Regions targeted by EWS-FLI are normally repressed and nucleosomal in primary endothelial cells. In tumor cells, however, bound regions are nucleosome depleted and harbor the chromatin signature of enhancers. We next demonstrated that through chimerism, EWS-FLI acquired the ability to alter chromatin. Expression of EWS-FLI results in nucleosome depletion at targeted sites, whereas silencing of EWS-FLI in tumor cells restored nucleosome occupancy. Thus, the EWS-FLI chimera acquired chromatin-altering activity, leading to mistargeting, chromatin disruption, and ultimately, transcriptional dysregulation.


Subject(s)
Chromatin/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Transcription, Genetic , Binding Sites/genetics , Cell Line, Tumor , Chimerism , Chromatin/metabolism , Endothelial Cells/metabolism , Enhancer Elements, Genetic , Gene Silencing , Humans , Microfilament Proteins/metabolism , Microsatellite Repeats , Neoplasms/genetics , Neoplasms/metabolism , Nucleotide Motifs , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Trans-Activators , Translocation, Genetic
19.
Genome Res ; 21(10): 1757-67, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21750106

ABSTRACT

The human body contains thousands of unique cell types, each with specialized functions. Cell identity is governed in large part by gene transcription programs, which are determined by regulatory elements encoded in DNA. To identify regulatory elements active in seven cell lines representative of diverse human cell types, we used DNase-seq and FAIRE-seq (Formaldehyde Assisted Isolation of Regulatory Elements) to map "open chromatin." Over 870,000 DNaseI or FAIRE sites, which correspond tightly to nucleosome-depleted regions, were identified across the seven cell lines, covering nearly 9% of the genome. The combination of DNaseI and FAIRE is more effective than either assay alone in identifying likely regulatory elements, as judged by coincidence with transcription factor binding locations determined in the same cells. Open chromatin common to all seven cell types tended to be at or near transcription start sites and to be coincident with CTCF binding sites, while open chromatin sites found in only one cell type were typically located away from transcription start sites and contained DNA motifs recognized by regulators of cell-type identity. We show that open chromatin regions bound by CTCF are potent insulators. We identified clusters of open regulatory elements (COREs) that were physically near each other and whose appearance was coordinated among one or more cell types. Gene expression and RNA Pol II binding data support the hypothesis that COREs control gene activity required for the maintenance of cell-type identity. This publicly available atlas of regulatory elements may prove valuable in identifying noncoding DNA sequence variants that are causally linked to human disease.


Subject(s)
Chromatin/metabolism , Chromosome Mapping , Regulatory Elements, Transcriptional , Sequence Analysis, DNA/methods , Base Sequence , Binding Sites , CCCTC-Binding Factor , Cell Differentiation/genetics , Cell Line , Gene Expression Regulation , Humans , Protein Binding , Repressor Proteins/metabolism , Transcription, Genetic , Transcriptional Activation
20.
J Virol ; 87(21): 11831-42, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23986576

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic gammaherpesvirus which establishes latent infection in endothelial and B cells, as well as in primary effusion lymphoma (PEL). During latency, the viral genome exists as a circular DNA minichromosome (episome) and is packaged into chromatin analogous to human chromosomes. Only a small subset of promoters, those which drive latent RNAs, are active in latent episomes. In general, nucleosome depletion ("open chromatin") is a hallmark of eukaryotic regulatory elements such as promoters and transcriptional enhancers or insulators. We applied formaldehyde-assisted isolation of regulatory elements (FAIRE) followed by next-generation sequencing to identify regulatory elements in the KSHV genome and integrated these data with previously identified locations of histone modifications, RNA polymerase II occupancy, and CTCF binding sites. We found that (i) regions of open chromatin were not restricted to the transcriptionally defined latent loci; (ii) open chromatin was adjacent to regions harboring activating histone modifications, even at transcriptionally inactive loci; and (iii) CTCF binding sites fell within regions of open chromatin with few exceptions, including the constitutive LANA promoter and the vIL6 promoter. FAIRE-identified nucleosome depletion was similar among B and endothelial cell lineages, suggesting a common viral genome architecture in all forms of latency.


Subject(s)
Chromatin/metabolism , DNA, Viral/metabolism , Herpesvirus 8, Human/physiology , Virus Latency , B-Lymphocytes/virology , Cell Line , Chromatin Immunoprecipitation , Endothelial Cells/virology , Gene Expression Profiling , Herpesvirus 8, Human/genetics , Histones/metabolism , Humans , Microarray Analysis , Protein Binding , Regulatory Elements, Transcriptional , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL