Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Adv Sci (Weinh) ; 11(16): e2303379, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38380561

RESUMO

Patient-Derived Organoids (PDO) and Xenografts (PDX) are the current gold standards for patient-derived models of cancer (PDMC). Nevertheless, how patient tumor cells evolve in these models and the impact on drug response remains unclear. Herein, the transcriptomic and chromatin accessibility landscapes of matched colorectal cancer (CRC) PDO, PDX, PDO-derived PDX (PDOX), and original patient tumors (PT) are compared. Two major remodeling axes are discovered. The first axis delineates PDMC from PT, and the second axis distinguishes PDX and PDO. PDOX are more similar to PDX than PDO, indicating the growth environment is a driving force for chromatin adaptation. Transcription factors (TF) that differentially bind to open chromatins between matched PDO and PDOX are identified. Among them, KLF14 and EGR2 footprints are enriched in PDOX relative to matched PDO, and silencing of KLF14 or EGR2 promoted tumor growth. Furthermore, EPHA4, a shared downstream target gene of KLF14 and EGR2, altered tumor sensitivity to MEK inhibitor treatment. Altogether, patient-derived CRC cells undergo both common and distinct chromatin remodeling in PDO and PDX/PDOX, driven largely by their respective microenvironments, which results in differences in growth and drug sensitivity and needs to be taken into consideration when interpreting their ability to predict clinical outcome.


Assuntos
Montagem e Desmontagem da Cromatina , Neoplasias Colorretais , Organoides , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Humanos , Montagem e Desmontagem da Cromatina/genética , Camundongos , Animais , Organoides/metabolismo , Modelos Animais de Doenças
2.
mBio ; 15(1): e0244423, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38059622

RESUMO

IMPORTANCE: Epstein-Barr virus has evolved with its human host leading to an intimate relationship where infection of antibody-producing B cells mimics the process by which these cells normally recognize foreign antigens and become activated. Virtually everyone in the world is infected by adulthood and controls this virus pushing it into life-long latency. However, immune-suppressed individuals are at high risk for EBV+ cancers. Here, we isolated B cells from tonsils and compare the underlying molecular genetic differences between these cells and those infected with EBV. We find similar regulatory mechanism for expression of an important cellular protein that enables B cells to survive in lymphoid tissue. These findings link an underlying relationship at the molecular level between EBV-infected B cells in vitro with normally activated B cells in vivo. Our studies also characterize the role of a key viral control mechanism for B cell survival involved in long-term infection.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Proteínas Proto-Oncogênicas c-bcl-2 , Adulto , Humanos , Cromatina , Antígenos Nucleares do Vírus Epstein-Barr , Centro Germinativo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Latência Viral , Proteínas Proto-Oncogênicas c-bcl-2/genética
3.
Dev Cell ; 57(3): 310-328.e9, 2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-35134344

RESUMO

Oncogenic Kras induces a hyper-proliferative state that permits cells to progress to neoplasms in diverse epithelial tissues. Depending on the cell of origin, this also involves lineage transformation. Although a multitude of downstream factors have been implicated in these processes, the precise chronology of molecular events controlling them remains elusive. Using mouse models, primary human tissues, and cell lines, we show that, in Kras-mutant alveolar type II cells (AEC2), FOSL1-based AP-1 factor guides the mSWI/SNF complex to increase chromatin accessibility at genomic loci controlling the expression of genes necessary for neoplastic transformation. We identified two orthogonal processes in Kras-mutant distal airway club cells. The first promoted their transdifferentiation into an AEC2-like state through NKX2.1, and the second controlled oncogenic transformation through the AP-1 complex. Our results suggest that neoplasms retain an epigenetic memory of their cell of origin through cell-type-specific transcription factors. Our analysis showed that a cross-tissue-conserved AP-1-dependent chromatin remodeling program regulates carcinogenesis.


Assuntos
Plasticidade Celular/genética , Epigênese Genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Oncogenes , Proteínas Proto-Oncogênicas p21(ras)/genética , Células Epiteliais Alveolares/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Proliferação de Células/genética , Epigenoma , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Mutantes/metabolismo , Mutação/genética , Neoplasias/patologia , Nucleossomos/metabolismo , Especificidade de Órgãos , Proteínas Proto-Oncogênicas c-fos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Fator de Transcrição AP-1/metabolismo
4.
Genes Dis ; 8(2): 203-214, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33997167

RESUMO

Colorectal cancer is a leading cause of cancer deaths. Most colorectal cancer patients eventually develop chemoresistance to the current standard-of-care therapies. Here, we used patient-derived colorectal cancer organoids to demonstrate that resistant tumor cells undergo significant chromatin changes in response to oxaliplatin treatment. Integrated transcriptomic and chromatin accessibility analyses using ATAC-Seq and RNA-Seq identified a group of genes associated with significantly increased chromatin accessibility and upregulated gene expression. CRISPR/Cas9 silencing of fibroblast growth factor receptor 1 (FGFR1) and oxytocin receptor (OXTR) helped overcome oxaliplatin resistance. Similarly, treatment with oxaliplatin in combination with an FGFR1 inhibitor (PD166866) or an antagonist of OXTR (L-368,899) suppressed chemoresistant organoids. However, oxaliplatin treatment did not activate either FGFR1 or OXTR expression in another resistant organoid, suggesting that chromatin accessibility changes are patient-specific. The use of patient-derived cancer organoids in combination with transcriptomic and chromatin profiling may lead to precision treatments to overcome chemoresistance in colorectal cancer.

5.
Cell Rep ; 29(4): 889-903.e10, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31644911

RESUMO

Notwithstanding the positive clinical impact of endocrine therapies in estrogen receptor-alpha (ERα)-positive breast cancer, de novo and acquired resistance limits the therapeutic lifespan of existing drugs. Taking the position that resistance is nearly inevitable, we undertook a study to identify and exploit targetable vulnerabilities that were manifest in endocrine therapy-resistant disease. Using cellular and mouse models of endocrine therapy-sensitive and endocrine therapy-resistant breast cancer, together with contemporary discovery platforms, we identified a targetable pathway that is composed of the transcription factors FOXA1 and GRHL2, a coregulated target gene, the membrane receptor LYPD3, and the LYPD3 ligand, AGR2. Inhibition of the activity of this pathway using blocking antibodies directed against LYPD3 or AGR2 inhibits the growth of endocrine therapy-resistant tumors in mice, providing the rationale for near-term clinical development of humanized antibodies directed against these proteins.


Assuntos
Fator 3-alfa Nuclear de Hepatócito/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Fatores de Transcrição/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Moléculas de Adesão Celular/imunologia , Moléculas de Adesão Celular/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio/genética , Feminino , Proteínas Ligadas por GPI/imunologia , Proteínas Ligadas por GPI/metabolismo , Humanos , Células MCF-7 , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Camundongos , Mucoproteínas/imunologia , Mucoproteínas/metabolismo , Proteínas Oncogênicas/imunologia , Proteínas Oncogênicas/metabolismo
6.
J Biol Chem ; 293(41): 15790-15800, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30093408

RESUMO

Interleukin (IL)-1ß plays a critical role in IL-6ß- and transforming growth factor ß (TGFß)-initiated Th17 differentiation and induction of Th17-mediated autoimmunity. However, the means by which IL-1 regulates various aspects of Th17 development remain poorly understood. We recently reported that IL-1ß enhances STAT3 phosphorylation via NF-κB-mediated repression of SOCS3 to facilitate Il17 transcription and Th17 differentiation, identifying an effect of IL-1 signaling on proximal events of STAT3 signaling. Here, we show that IL-1ß promotes STAT3 binding to key cis-elements that control IL-17 expression. Additionally, we demonstrate that the IL-1-induced NF-κB factor RelA directly regulates the Il17a/f loci in cooperation with STAT3. Our findings reveal that IL-1 impacts both proximal signaling events and downstream interactions between transcription factors and cis-regulatory elements to promote Il17a/f transcription and Th17 differentiation.


Assuntos
Interleucina-17/metabolismo , Receptores Tipo II de Interleucina-1/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , DNA/química , DNA/genética , Interleucina-17/genética , Camundongos Endogâmicos C57BL , Sequências Reguladoras de Ácido Nucleico/genética , Fator de Transcrição STAT3/genética , Transdução de Sinais/fisiologia , Células Th17 , Fator de Transcrição RelA/genética , Ativação Transcricional
7.
Genome Res ; 28(9): 1272-1284, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30097539

RESUMO

Glucocorticoids are potent steroid hormones that regulate immunity and metabolism by activating the transcription factor (TF) activity of glucocorticoid receptor (GR). Previous models have proposed that DNA binding motifs and sites of chromatin accessibility predetermine GR binding and activity. However, there are vast excesses of both features relative to the number of GR binding sites. Thus, these features alone are unlikely to account for the specificity of GR binding and activity. To identify genomic and epigenetic contributions to GR binding specificity and the downstream changes resultant from GR binding, we performed hundreds of genome-wide measurements of TF binding, epigenetic state, and gene expression across a 12-h time course of glucocorticoid exposure. We found that glucocorticoid treatment induces GR to bind to nearly all pre-established enhancers within minutes. However, GR binds to only a small fraction of the set of accessible sites that lack enhancer marks. Once GR is bound to enhancers, a combination of enhancer motif composition and interactions between enhancers then determines the strength and persistence of GR binding, which consequently correlates with dramatic shifts in enhancer activation. Over the course of several hours, highly coordinated changes in TF binding and histone modification occupancy occur specifically within enhancers, and these changes correlate with changes in the expression of nearby genes. Following GR binding, changes in the binding of other TFs precede changes in chromatin accessibility, suggesting that other TFs are also sensitive to genomic features beyond that of accessibility.


Assuntos
Elementos Facilitadores Genéticos , Código das Histonas , Motivos de Nucleotídeos , Receptores de Glucocorticoides/metabolismo , Ativação Transcricional , Linhagem Celular Tumoral , Epigênese Genética , Humanos , Ligação Proteica , Fatores de Transcrição/metabolismo
8.
Genome Biol Evol ; 10(3): 826-839, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29608722

RESUMO

Humans experience higher rates of age-associated diseases than our closest living evolutionary relatives, chimpanzees. Environmental factors can explain many of these increases in disease risk, but species-specific genetic changes can also play a role. Alleles that confer increased disease susceptibility later in life can persist in a population in the absence of selective pressure if those changes confer positive adaptation early in life. One age-associated disease that disproportionately affects humans compared with chimpanzees is epithelial cancer. Here, we explored genetic differences between humans and chimpanzees in a well-defined experimental assay that mimics gene expression changes that happen during cancer progression: A fibroblast serum challenge. We used this assay with fibroblasts isolated from humans and chimpanzees to explore species-specific differences in gene expression and chromatin state with RNA-Seq and DNase-Seq. Our data reveal that human fibroblasts increase expression of genes associated with wound healing and cancer pathways; in contrast, chimpanzee gene expression changes are not concentrated around particular functional categories. Chromatin accessibility dramatically increases in human fibroblasts, yet decreases in chimpanzee cells during the serum response. Many regions of opening and closing chromatin are in close proximity to genes encoding transcription factors or genes involved in wound healing processes, further supporting the link between changes in activity of regulatory elements and changes in gene expression. Together, these expression and open chromatin data show that humans and chimpanzees have dramatically different responses to the same physiological stressor, and how a core physiological process can evolve quickly over relatively short evolutionary time scales.


Assuntos
Cromatina/genética , Evolução Molecular , Variação Genética/genética , Sequências Reguladoras de Ácido Nucleico/genética , Animais , Regulação da Expressão Gênica/genética , Humanos , Pan troglodytes/sangue , Pan troglodytes/genética , Regiões Promotoras Genéticas , Especificidade da Espécie , Fatores de Transcrição/genética
9.
Mamm Genome ; 29(1-2): 153-167, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29429127

RESUMO

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.


Assuntos
Dano ao DNA/efeitos dos fármacos , Epigênese Genética , Transcrição Gênica/genética , Transcriptoma/genética , Animais , Butadienos/toxicidade , Carcinógenos/toxicidade , Cromatina/efeitos dos fármacos , Histonas/genética , Fígado/efeitos dos fármacos , Fígado/patologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Testes de Mutagenicidade , Especificidade de Órgãos/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
10.
Environ Health Perspect ; 125(10): 107006, 2017 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-29038090

RESUMO

BACKGROUND: The damaging effects of exposure to environmental toxicants differentially affect genetically distinct individuals, but the mechanisms contributing to these differences are poorly understood. Genetic variation affects the establishment of the gene regulatory landscape and thus gene expression, and we hypothesized that this contributes to the observed heterogeneity in individual responses to exogenous cellular insults. OBJECTIVES: We performed an in vivo study of how genetic variation and chromatin organization may dictate susceptibility to DNA damage, and influence the cellular response to such damage, caused by an environmental toxicant. MATERIALS AND METHODS: We measured DNA damage, messenger RNA (mRNA) and microRNA (miRNA) expression, and genome-wide chromatin accessibility in lung tissue from two genetically divergent inbred mouse strains, C57BL/6J and CAST/EiJ, both in unexposed mice and in mice exposed to a model DNA-damaging chemical, 1,3-butadiene. RESULTS: Our results showed that unexposed CAST/EiJ and C57BL/6J mice have very different chromatin organization and transcription profiles in the lung. Importantly, in unexposed CAST/EiJ mice, which acquired relatively less 1,3-butadiene-induced DNA damage, we observed increased transcription and a more accessible chromatin landscape around genes involved in detoxification pathways. Upon chemical exposure, chromatin was significantly remodeled in the lung of C57BL/6J mice, a strain that acquired higher levels of 1,3-butadiene-induced DNA damage, around the same genes, ultimately resembling the molecular profile of CAST/EiJ. CONCLUSIONS: These results suggest that strain-specific changes in chromatin and transcription in response to chemical exposure lead to a "compensation" for underlying genetic-driven interindividual differences in the baseline chromatin and transcriptional state. This work represents an example of how chemical and environmental exposures can be evaluated to better understand gene-by-environment interactions, and it demonstrates the important role of chromatin response in transcriptomic changes and, potentially, in deleterious effects of exposure. https://doi.org/10.1289/EHP1937.


Assuntos
Poluentes Atmosféricos/toxicidade , Butadienos/toxicidade , Dano ao DNA , Transcrição Gênica/efeitos dos fármacos , Animais , Cromatina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos
11.
Artigo em Inglês | MEDLINE | ID: mdl-27087856

RESUMO

BACKGROUND: Small molecule inhibitors of histone deacetylases (HDACi) hold promise as anticancer agents for particular malignancies. However, clinical use is often confounded by toxicity, perhaps due to indiscriminate hyperacetylation of cellular proteins. Therefore, elucidating the mechanisms by which HDACi trigger differentiation, cell cycle arrest, or apoptosis of cancer cells could inform development of more targeted therapies. We used the myelogenous leukemia line K562 as a model of HDACi-induced differentiation to investigate chromatin accessibility (DNase-seq) and expression (RNA-seq) changes associated with this process. RESULTS: We identified several thousand specific regulatory elements [~10 % of total DNase I-hypersensitive (DHS) sites] that become significantly more or less accessible with sodium butyrate or suberanilohydroxamic acid treatment. Most of the differential DHS sites display hallmarks of enhancers, including being enriched for non-promoter regions, associating with nearby gene expression changes, and increasing luciferase reporter expression in K562 cells. Differential DHS sites were enriched for key hematopoietic lineage transcription factor motifs, including SPI1 (PU.1), a known pioneer factor. We found PU.1 increases binding at opened DHS sites with HDACi treatment by ChIP-seq, but PU.1 knockdown by shRNA fails to block the chromatin accessibility and expression changes. A machine-learning approach indicates H3K27me3 initially marks PU.1-bound sites that open with HDACi treatment, suggesting these sites are epigenetically poised. CONCLUSIONS: We find HDACi treatment of K562 cells results in site-specific chromatin remodeling at epigenetically poised regulatory elements. PU.1 shows evidence of a pioneer role in this process by marking poised enhancers but is not required for transcriptional activation.

12.
Nucleic Acids Res ; 44(7): 3082-94, 2016 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-26673704

RESUMO

Higher order chromatin structure establishes domains that organize the genome and coordinate gene expression. However, the molecular mechanisms controlling transcription of individual loci within a topological domain (TAD) are not fully understood. The cystic fibrosis transmembrane conductance regulator (CFTR) gene provides a paradigm for investigating these mechanisms.CFTR occupies a TAD bordered by CTCF/cohesin binding sites within which are cell-type-selective cis-regulatory elements for the locus. We showed previously that intronic and extragenic enhancers, when occupied by specific transcription factors, are recruited to the CFTR promoter by a looping mechanism to drive gene expression. Here we use a combination of CRISPR/Cas9 editing of cis-regulatory elements and siRNA-mediated depletion of architectural proteins to determine the relative contribution of structural elements and enhancers to the higher order structure and expression of the CFTR locus. We found the boundaries of the CFTRTAD are conserved among diverse cell types and are dependent on CTCF and cohesin complex. Removal of an upstream CTCF-binding insulator alters the interaction profile, but has little effect on CFTR expression. Within the TAD, intronic enhancers recruit cell-type selective transcription factors and deletion of a pivotal enhancer element dramatically decreases CFTR expression, but has minor effect on its 3D structure.


Assuntos
Cromatina/química , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Elementos Isolantes , Fator de Ligação a CCCTC , Células CACO-2 , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Células Cultivadas , Proteínas Cromossômicas não Histona/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Loci Gênicos , Humanos , Proteínas Repressoras/metabolismo , Coesinas
13.
Hum Mol Genet ; 24(19): 5433-50, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26206884

RESUMO

SOX10 is required for melanocyte development and maintenance, and has been linked to melanoma initiation and progression. However, the molecular mechanisms by which SOX10 guides the appropriate gene expression programs necessary to promote the melanocyte lineage are not fully understood. Here we employ genetic and epigenomic analysis approaches to uncover novel genomic targets and previously unappreciated molecular roles of SOX10 in melanocytes. Through global analysis of SOX10-binding sites and epigenetic characteristics of chromatin states, we uncover an extensive catalog of SOX10 targets genome-wide. Our findings reveal that SOX10 predominantly engages 'open' chromatin regions and binds to distal regulatory elements, including novel and previously known melanocyte enhancers. Integrated chromatin occupancy and transcriptome analysis suggest a role for SOX10 in both transcriptional activation and repression to regulate functionally distinct classes of genes. We demonstrate that distinct epigenetic signatures and cis-regulatory sequence motifs predicted to bind putative co-regulatory transcription factors define SOX10-activated and SOX10-repressed target genes. Collectively, these findings uncover a central role of SOX10 as a global regulator of gene expression in the melanocyte lineage by targeting diverse regulatory pathways.


Assuntos
Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Melanócitos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Fatores de Transcrição SOXE/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Cromatina/genética , Cromatina/metabolismo , Epigenômica/métodos , Melanócitos/citologia , Camundongos , Fatores de Transcrição SOXE/química , Fatores de Transcrição SOXE/genética
14.
PLoS Genet ; 11(1): e1004885, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25569532

RESUMO

Cellular stresses activate the tumor suppressor p53 protein leading to selective binding to DNA response elements (REs) and gene transactivation from a large pool of potential p53 REs (p53REs). To elucidate how p53RE sequences and local chromatin context interact to affect p53 binding and gene transactivation, we mapped genome-wide binding localizations of p53 and H3K4me3 in untreated and doxorubicin (DXR)-treated human lymphoblastoid cells. We examined the relationships among p53 occupancy, gene expression, H3K4me3, chromatin accessibility (DNase 1 hypersensitivity, DHS), ENCODE chromatin states, p53RE sequence, and evolutionary conservation. We observed that the inducible expression of p53-regulated genes was associated with the steady-state chromatin status of the cell. Most highly inducible p53-regulated genes were suppressed at baseline and marked by repressive histone modifications or displayed CTCF binding. Comparison of p53RE sequences residing in different chromatin contexts demonstrated that weaker p53REs resided in open promoters, while stronger p53REs were located within enhancers and repressed chromatin. p53 occupancy was strongly correlated with similarity of the target DNA sequences to the p53RE consensus, but surprisingly, inversely correlated with pre-existing nucleosome accessibility (DHS) and evolutionary conservation at the p53RE. Occupancy by p53 of REs that overlapped transposable element (TE) repeats was significantly higher (p<10-7) and correlated with stronger p53RE sequences (p<10-110) relative to nonTE-associated p53REs, particularly for MLT1H, LTR10B, and Mer61 TEs. However, binding at these elements was generally not associated with transactivation of adjacent genes. Occupied p53REs located in L2-like TEs were unique in displaying highly negative PhyloP scores (predicted fast-evolving) and being associated with altered H3K4me3 and DHS levels. These results underscore the systematic interaction between chromatin status and p53RE context in the induced transactivation response. This p53 regulated response appears to have been tuned via evolutionary processes that may have led to repression and/or utilization of p53REs originating from primate-specific transposon elements.


Assuntos
Cromatina/genética , Elementos de Resposta/genética , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Animais , Sítios de Ligação , Cromatina/efeitos dos fármacos , Estruturas Cromossômicas/efeitos dos fármacos , Estruturas Cromossômicas/genética , Elementos de DNA Transponíveis , Doxorrubicina/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase , Humanos , Nucleossomos/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteína Supressora de Tumor p53/metabolismo
15.
Mol Hum Reprod ; 20(12): 1198-207, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25180270

RESUMO

The epithelium lining the epididymis in the male reproductive tract maintains a luminal environment that promotes sperm cell maturation. This process is dependent on the coordinated expression of many genes that encode proteins with a role in epithelial transport. We previously generated genome-wide maps of open chromatin in primary human epididymis epithelial (HEE) cells to identify potential regulatory elements controlling coordinated gene expression in the epididymis epithelium. Subsequent in silico analysis identified transcription factor-binding sites (TFBS) that were over-represented in the HEE open chromatin, including the motif for paired box 2 (PAX2). PAX2 is a critical transcriptional regulator of urogenital tract development, which has been well studied in the kidney but is unexplored in the epididymis. Due to the limited lifespan of primary HEE cells in culture, we investigated the role of PAX2 in an immortalized HEE cell line (REP). First, REP cells were evaluated by DNase I digestion followed by high-throughput sequencing and the PAX2-binding motif was again identified as an over-represented TFBS within intergenic open chromatin, though on fewer chromosomes than in the primary HEE cells. To identify PAX2-target genes in REP cells, RNA-seq analysis was performed after siRNA-mediated depletion of PAX2 and compared with that with a non-targeting siRNA. In response to PAX2-repression, 3135 transcripts were differentially expressed (1333 up-regulated and 1802 down-regulated). Novel PAX2 targets included multiple genes encoding proteins with predicted functions in the epididymis epithelium.


Assuntos
Montagem e Desmontagem da Cromatina , Epididimo/metabolismo , Células Epiteliais/metabolismo , Redes Reguladoras de Genes , Fator de Transcrição PAX2/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Linhagem Celular , Epididimo/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Fator de Transcrição PAX2/genética , Interferência de RNA , Transcrição Gênica , Transfecção
16.
PLoS One ; 9(9): e107156, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25198066

RESUMO

Despite an emerging understanding of the genetic alterations giving rise to various tumors, the mechanisms whereby most oncogenes are overexpressed remain unclear. Here we have utilized an integrated approach of genomewide regulatory element mapping via DNase-seq followed by conventional reporter assays and transcription factor binding site discovery to characterize the transcriptional regulation of the medulloblastoma oncogene Orthodenticle Homeobox 2 (OTX2). Through these studies we have revealed that OTX2 is differentially regulated in medulloblastoma at the level of chromatin accessibility, which is in part mediated by DNA methylation. In cell lines exhibiting chromatin accessibility of OTX2 regulatory regions, we found that autoregulation maintains OTX2 expression. Comparison of medulloblastoma regulatory elements with those of the developing brain reveals that these tumors engage a developmental regulatory program to drive OTX2 transcription. Finally, we have identified a transcriptional regulatory element mediating retinoid-induced OTX2 repression in these tumors. This work characterizes for the first time the mechanisms of OTX2 overexpression in medulloblastoma. Furthermore, this study establishes proof of principle for applying ENCODE datasets towards the characterization of upstream trans-acting factors mediating expression of individual genes.


Assuntos
Neoplasias Cerebelares/genética , Cromatina/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Meduloblastoma/genética , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo , Retinoides/farmacologia , Neoplasias Cerebelares/metabolismo , Neoplasias Cerebelares/patologia , Cromatina/química , Imunoprecipitação da Cromatina , Estruturas Cromossômicas/genética , Metilação de DNA/efeitos dos fármacos , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patologia , Regiões Promotoras Genéticas/genética , Elementos Reguladores de Transcrição/genética , Transativadores/genética , Células Tumorais Cultivadas
17.
Epigenetics ; 9(6): 842-50, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24670287

RESUMO

Notch intercellular signaling is critical for diverse developmental pathways and for homeostasis in various types of stem cells and progenitor cells. Because Notch gene products need to be precisely regulated spatially and temporally, epigenetics is likely to help control expression of Notch signaling genes. Reduced representation bisulfite sequencing (RRBS) indicated significant hypomethylation in myoblasts, myotubes, and skeletal muscle vs. many nonmuscle samples at intragenic or intergenic regions of the following Notch receptor or ligand genes: NOTCH1, NOTCH2, JAG2, and DLL1. An enzymatic assay of sites in or near these genes revealed unusually high enrichment of 5-hydroxymethylcytosine (up to 81%) in skeletal muscle, heart, and cerebellum. Epigenetics studies and gene expression profiles suggest that hypomethylation and/or hydroxymethylation help control expression of these genes in heart, brain, myoblasts, myotubes, and within skeletal muscle myofibers. Such regulation could promote cell renewal, cell maintenance, homeostasis, and a poised state for repair of tissue damage.


Assuntos
Cerebelo/metabolismo , Citosina/análogos & derivados , Metilação de DNA , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Receptores Notch/genética , 5-Metilcitosina/análogos & derivados , Idoso , Idoso de 80 Anos ou mais , Cromatina/metabolismo , Citosina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Feminino , Histonas/metabolismo , Humanos , Masculino , Oxigenases de Função Mista , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/metabolismo , Especificidade de Órgãos , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/metabolismo , Receptores Notch/metabolismo
18.
Mol Cell ; 52(1): 25-36, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-24076218

RESUMO

Most human transcription factors bind a small subset of potential genomic sites and often use different subsets in different cell types. To identify mechanisms that govern cell-type-specific transcription factor binding, we used an integrative approach to study estrogen receptor α (ER). We found that ER exhibits two distinct modes of binding. Shared sites, bound in multiple cell types, are characterized by high-affinity estrogen response elements (EREs), inaccessible chromatin, and a lack of DNA methylation, while cell-specific sites are characterized by a lack of EREs, co-occurrence with other transcription factors, and cell-type-specific chromatin accessibility and DNA methylation. These observations enabled accurate quantitative models of ER binding that suggest tethering of ER to one-third of cell-specific sites. The distinct properties of cell-specific binding were also observed with glucocorticoid receptor and for ER in primary mouse tissues, representing an elegant genomic encoding scheme for generating cell-type-specific gene regulation.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Sequência Conservada , Metilação de DNA , Estradiol/farmacologia , Receptor alfa de Estrogênio/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Estrogênios/farmacologia , Evolução Molecular , Regulação da Expressão Gênica , Humanos , Camundongos , Modelos Biológicos , Regiões Promotoras Genéticas/efeitos dos fármacos , Interferência de RNA , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Elementos de Resposta , Termodinâmica , Fatores de Transcrição/genética , Transfecção
19.
Biol Reprod ; 89(4): 104, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24006278

RESUMO

The epithelium lining the epididymis has a pivotal role in ensuring a luminal environment that can support normal sperm maturation. Many of the individual genes that encode proteins involved in establishing the epididymal luminal fluid are well characterized. They include ion channels, ion exchangers, transporters, and solute carriers. However, the molecular mechanisms that coordinate expression of these genes and modulate their activities in response to biological stimuli are less well understood. To identify cis-regulatory elements for genes expressed in human epididymis epithelial cells, we generated genome-wide maps of open chromatin by DNase-seq. This analysis identified 33,542 epididymis-selective DNase I hypersensitive sites (DHS), which were not evident in five cell types of different lineages. Identification of genes with epididymis-selective DHS at their promoters revealed gene pathways that are active in immature epididymis epithelial cells. These include processes correlating with epithelial function and also others with specific roles in the epididymis, including retinol metabolism and ascorbate and aldarate metabolism. Peaks of epididymis-selective chromatin were seen in the androgen receptor gene and the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which has a critical role in regulating ion transport across the epididymis epithelium. In silico prediction of transcription factor binding sites that were overrepresented in epididymis-selective DHS identified epithelial transcription factors, including ELF5 and ELF3, the androgen receptor, Pax2, and Sox9, as components of epididymis transcriptional networks. Active genes, which are targets of each transcription factor, reveal important biological processes in the epididymis epithelium.


Assuntos
Montagem e Desmontagem da Cromatina , Epididimo/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Infertilidade Masculina/genética , Espermatogênese , Células Cultivadas , Mapeamento Cromossômico , Biologia Computacional , DNA Intergênico , Epididimo/citologia , Epididimo/crescimento & desenvolvimento , Epididimo/fisiopatologia , Células Epiteliais/citologia , Sistemas Inteligentes , Feto/citologia , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Infertilidade Masculina/metabolismo , Infertilidade Masculina/fisiopatologia , Masculino , Nucleossomos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Regiões Promotoras Genéticas
20.
Genome Res ; 23(6): 917-27, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23547170

RESUMO

Histone modifications are now well-established mediators of transcriptional programs that distinguish cell states. However, the kinetics of histone modification and their role in mediating rapid, signal-responsive gene expression changes has been little studied on a genome-wide scale. Vascular endothelial growth factor A (VEGFA), a major regulator of angiogenesis, triggers changes in transcriptional activity of human umbilical vein endothelial cells (HUVECs). Here, we used chromatin immunoprecipitation followed by high-throughput sequencing (ChIP-seq) to measure genome-wide changes in histone H3 acetylation at lysine 27 (H3K27ac), a marker of active enhancers, in unstimulated HUVECs and HUVECs stimulated with VEGFA for 1, 4, and 12 h. We show that sites with the greatest H3K27ac change upon stimulation were associated tightly with EP300, a histone acetyltransferase. Using the variation of H3K27ac as a novel epigenetic signature, we identified transcriptional regulatory elements that are functionally linked to angiogenesis, participate in rapid VEGFA-stimulated changes in chromatin conformation, and mediate VEGFA-induced transcriptional responses. Dynamic H3K27ac deposition and associated changes in chromatin conformation required EP300 activity instead of altered nucleosome occupancy or changes in DNase I hypersensitivity. EP300 activity was also required for a subset of dynamic H3K27ac sites to loop into proximity of promoters. Our study identified thousands of endothelial, VEGFA-responsive enhancers, demonstrating that an epigenetic signature based on the variation of a chromatin feature is a productive approach to define signal-responsive genomic elements. Further, our study implicates global epigenetic modifications in rapid, signal-responsive transcriptional regulation.


Assuntos
Proteína p300 Associada a E1A/metabolismo , Células Endoteliais/metabolismo , Elementos Facilitadores Genéticos , Histonas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Acetilação , Sítios de Ligação , Cromatina/genética , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina , Análise por Conglomerados , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Motivos de Nucleotídeos , Ligação Proteica , Elementos de Resposta , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA