Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Cell ; 159(6): 1249-51, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25480289

RESUMO

A high-resolution look at where histones touch DNA reveals a surprisingly intricate, dynamic, and modular nucleosome. Three advances in the study by Rhee et al. include unexpected interactions between the H3 tail and linker DNA, new evidence for existence of subnucleosomal particles, and asymmetric patterns of histone modification within a single nucleosome that correspond to the direction of transcription.


Assuntos
Nucleossomos/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transcrição Gênica
2.
Cell ; 133(5): 763-5, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18510918

RESUMO

The histone acetyltransferase MOF (males-absent-on-the-first) is required for the regulation of X chromosome gene dosage compensation in Drosophila males. In this issue, Kind et al. (2008) show that MOF is also found on autosomes and that it has two modes of binding: one in males for X chromosome dosage compensation and the other in both sexes for X chromosome and autosomal gene regulation independent of dosage compensation.


Assuntos
Mecanismo Genético de Compensação de Dose , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Proteínas Nucleares/metabolismo , Animais , Masculino
3.
Genes Dev ; 29(13): 1393-402, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26104467

RESUMO

Chromosome translocations are well-established hallmarks of cancer cells and often occur at nonrandom sites in the genome. The molecular features that define recurrent chromosome breakpoints are largely unknown. Using a combination of bioinformatics, biochemical analysis, and cell-based assays, we identify here specific histone modifications as facilitators of chromosome breakage and translocations. We show enrichment of several histone modifications over clinically relevant translocation-prone genome regions. Experimental modulation of histone marks sensitizes genome regions to breakage by endonuclease challenge or irradiation and promotes formation of chromosome translocations of endogenous gene loci. Our results demonstrate that histone modifications predispose genome regions to chromosome breakage and translocations.


Assuntos
Quebra Cromossômica , Genoma Humano/genética , Histonas/metabolismo , Translocação Genética , Linhagem Celular Tumoral , Biologia Computacional , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Endonucleases/metabolismo , Histonas/genética , Humanos , Linfoma Anaplásico de Células Grandes/fisiopatologia , Metilação
4.
Genes Dev ; 29(9): 948-60, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25934506

RESUMO

We developed a method for genome-wide mapping of DNA excision repair named XR-seq (excision repair sequencing). Human nucleotide excision repair generates two incisions surrounding the site of damage, creating an ∼30-mer. In XR-seq, this fragment is isolated and subjected to high-throughput sequencing. We used XR-seq to produce stranded, nucleotide-resolution maps of repair of two UV-induced DNA damages in human cells: cyclobutane pyrimidine dimers (CPDs) and (6-4) pyrimidine-pyrimidone photoproducts [(6-4)PPs]. In wild-type cells, CPD repair was highly associated with transcription, specifically with the template strand. Experiments in cells defective in either transcription-coupled excision repair or general excision repair isolated the contribution of each pathway to the overall repair pattern and showed that transcription-coupled repair of both photoproducts occurs exclusively on the template strand. XR-seq maps capture transcription-coupled repair at sites of divergent gene promoters and bidirectional enhancer RNA (eRNA) production at enhancers. XR-seq data also uncovered the repair characteristics and novel sequence preferences of CPDs and (6-4)PPs. XR-seq and the resulting repair maps will facilitate studies of the effects of genomic location, chromatin context, transcription, and replication on DNA repair in human cells.


Assuntos
Dano ao DNA/efeitos da radiação , Reparo do DNA/genética , Nucleotídeos/genética , Raios Ultravioleta , Linhagem Celular , Elementos Facilitadores Genéticos/genética , Estudo de Associação Genômica Ampla , Humanos , Regiões Promotoras Genéticas/genética , Dímeros de Pirimidina/genética , Transcrição Gênica/genética
5.
Mol Cell ; 51(6): 840-9, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-24011592

RESUMO

Nuclear pores associate with active protein-coding genes in yeast and have been implicated in transcriptional regulation. Here, we show that in addition to transcriptional regulation, key components of C. elegans nuclear pores are required for processing of a subset of small nucleolar RNAs (snoRNAs) and tRNAs transcribed by RNA polymerase (Pol) III. Chromatin immunoprecipitation of NPP-13 and NPP-3, two integral nuclear pore components, and importin-ß IMB-1 provides strong evidence that this requirement is direct. All three proteins associate specifically with tRNA and snoRNA genes undergoing Pol III transcription. These pore components bind immediately downstream of the Pol III preinitiation complex but are not required for Pol III recruitment. Instead, NPP-13 is required for cleavage of tRNA and snoRNA precursors into mature RNAs, whereas Pol II transcript processing occurs normally. Our data suggest that integral nuclear pore proteins act to coordinate transcription and processing of Pol III transcripts in C. elegans.


Assuntos
Caenorhabditis elegans/genética , Poro Nuclear/genética , RNA Polimerase III/genética , Transcrição Gênica , Animais , Proteínas de Caenorhabditis elegans/metabolismo , Carioferinas/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , RNA Polimerase III/metabolismo , RNA Nucleolar Pequeno/genética , RNA Nucleolar Pequeno/metabolismo , RNA de Transferência/genética , RNA de Transferência/metabolismo
6.
Genome Res ; 27(1): 75-86, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27979995

RESUMO

Nucleosomes have structural and regulatory functions in all eukaryotic DNA-templated processes. The position of nucleosomes on DNA and the stability of the underlying histone-DNA interactions affect the access of regulatory proteins to DNA. Both stability and position are regulated through DNA sequence, histone post-translational modifications, histone variants, chromatin remodelers, and transcription factors. Here, we explored the functional implications of nucleosome properties on gene expression and development in Caenorhabditis elegans embryos. We performed a time-course of micrococcal nuclease (MNase) digestion and measured the relative sensitivity or resistance of nucleosomes throughout the genome. Fragile nucleosomes were defined by nucleosomal DNA fragments that were recovered preferentially in early MNase-digestion time points. Nucleosome fragility was strongly and positively correlated with the AT content of the underlying DNA sequence. There was no correlation between promoter nucleosome fragility and the levels of histone modifications or histone variants. Genes with fragile nucleosomes in their promoters tended to be lowly expressed and expressed in a context-specific way, operating in neuronal response, the immune system, and stress response. In addition to DNA-encoded nucleosome fragility, we also found fragile nucleosomes at locations where we expected to find destabilized nucleosomes, for example, at transcription factor binding sites where nucleosomes compete with DNA-binding factors. Our data suggest that in C. elegans promoters, nucleosome fragility is in large part DNA-encoded and that it poises genes for future context-specific activation in response to environmental stress and developmental cues.


Assuntos
Proteínas de Ligação a DNA/genética , Histonas/genética , Nucleossomos/genética , Transcrição Gênica , Animais , Sítios de Ligação/efeitos dos fármacos , Caenorhabditis elegans/genética , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Histonas/metabolismo , Nuclease do Micrococo/farmacologia , Regiões Promotoras Genéticas , Fatores de Transcrição/genética
7.
Development ; 143(20): 3723-3732, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27802137

RESUMO

One of the key issues in studying transcriptional regulation during development is how to employ genome-wide assays that reveals sites of open chromatin and transcription factor binding to efficiently identify biologically relevant genes and enhancers. Analysis of Drosophila CNS midline cell development provides a useful system for studying transcriptional regulation at the genomic level due to a large, well-characterized set of midline-expressed genes and in vivo validated enhancers. In this study, FAIRE-seq on FACS-purified midline cells was performed and the midline FAIRE data were compared with whole-embryo FAIRE data. We find that regions of the genome with a strong midline FAIRE peak and weak whole-embryo FAIRE peak overlap with known midline enhancers and provide a useful predictive tool for enhancer identification. In a complementary analysis, we compared a large dataset of fragments that drive midline expression in vivo with the FAIRE data. Midline enhancer fragments with a midline FAIRE peak tend to be near midline-expressed genes, whereas midline enhancers without a midline FAIRE peak were often distant from midline-expressed genes and unlikely to drive midline transcription in vivo.


Assuntos
Cromatina/metabolismo , Proteínas de Drosophila/metabolismo , Animais , Animais Geneticamente Modificados , Cromatina/genética , Imunoprecipitação da Cromatina , Drosophila , Proteínas de Drosophila/genética , Elementos Facilitadores Genéticos/genética , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento/genética
8.
Development ; 143(3): 483-91, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26700680

RESUMO

ELT-2 is the major regulator of genes involved in differentiation, maintenance and function of C. elegans intestine from the early embryo to mature adult. elt-2 responds to overexpression of the GATA transcription factors END-1 and END-3, which specify the intestine, as well as to overexpression of the two GATA factors that are normally involved in intestinal differentiation, ELT-7 and ELT-2 itself. Little is known about the molecular mechanisms underlying these interactions, how ELT-2 levels are maintained throughout development or how such systems respond to developmental perturbations. Here, we analyse elt-2 gene regulation through transgenic reporter assays, ELT-2 ChIP and characterisation of in vitro DNA-protein interactions. Our results indicate that elt-2 is controlled by three discrete regulatory regions conserved between C. elegans and C. briggsae that span >4 kb of 5' flanking sequence. These regions are superficially interchangeable but have quantitatively different enhancer properties, and their combined activities indicate inter-region synergies. Their regulatory activity is mediated by a small number of conserved TGATAA sites that are largely interchangeable and interact with different endodermal GATA factors with only modest differences in affinity. The redundant molecular mechanism that forms the elt-2 regulatory network is robust and flexible, as loss of end-3 halves ELT-2 levels in the early embryo but levels fully recover by the time of hatching. When ELT-2 is expressed under the control of end-1 regulatory elements, in addition to its own endogenous promoter, it can replace the complete set of endoderm-specific GATA factors: END-1, END-3, ELT-7 and (the probably non-functional) ELT-4. Thus, in addition to controlling gene expression during differentiation, ELT-2 is capable of specifying the entire C. elegans endoderm.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/genética , Endoderma/embriologia , Endoderma/metabolismo , Fatores de Transcrição GATA/genética , Regulação da Expressão Gênica no Desenvolvimento , Região 5'-Flanqueadora/genética , Animais , Sequência de Bases , Proteínas de Caenorhabditis elegans/metabolismo , Diferenciação Celular/genética , Imunoprecipitação da Cromatina , Sequência Conservada , DNA/metabolismo , Fatores de Transcrição GATA/metabolismo , Redes Reguladoras de Genes , Mucosa Intestinal/metabolismo , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ligação Proteica/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica
9.
Proc Natl Acad Sci U S A ; 113(15): E2124-33, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27036006

RESUMO

We recently developed a high-resolution genome-wide assay for mapping DNA excision repair named eXcision Repair-sequencing (XR-seq) and have now used XR-seq to determine which regions of the genome are subject to repair very soon after UV exposure and which regions are repaired later. Over a time course, we measured repair of the UV-induced damage of cyclobutane pyrimidine dimers (CPDs) (at 1, 4, 8, 16, 24, and 48 h) and (6-4)pyrimidine-pyrimidone photoproducts [(6-4)PPs] (at 5 and 20 min and 1, 2, and 4 h) in normal human skin fibroblasts. Each type of damage has distinct repair kinetics. The (6-4)PPs are detected as early as 5 min after UV treatment, with the bulk of repair completed by 4 h. Repair of CPDs, which we previously showed is intimately coupled to transcription, is slower and in certain regions persists even 2 d after UV irradiation. We compared our results to the Encyclopedia of DNA Elements data regarding histone modifications, chromatin state, and transcription. For both damage types, and for both transcription-coupled and general excision repair, the earliest repair occurred preferentially in active and open chromatin states. Conversely, repair in regions classified as "heterochromatic" and "repressed" was relatively low at early time points, with repair persisting into the late time points. Damage that remains during DNA replication increases the risk for mutagenesis. Indeed, late-repaired regions are associated with a higher level of cancer-linked mutations. In summary, we show that XR-seq is a powerful approach for studying relationships among chromatin state, DNA repair, genome stability, mutagenesis, and carcinogenesis.


Assuntos
Cromatina/genética , Reparo do DNA/genética , Genoma Humano/genética , Raios Ultravioleta/efeitos adversos , Linhagem Celular , Humanos , Cinética , Melanoma/genética , Mutagênese/genética , Análise de Sequência de DNA/métodos
10.
Proc Natl Acad Sci U S A ; 113(41): 11507-11512, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27688757

RESUMO

Cisplatin is a major anticancer drug that kills cancer cells by damaging their DNA. Cancer cells cope with the drug by removal of the damages with nucleotide excision repair. We have developed methods to measure cisplatin adduct formation and its repair at single-nucleotide resolution. "Damage-seq" relies on the replication-blocking properties of the bulky base lesions to precisely map their location. "XR-seq" independently maps the removal of these damages by capturing and sequencing the excised oligomer released during repair. The damage and repair maps we generated reveal that damage distribution is essentially uniform and is dictated mostly by the underlying sequence. In contrast, cisplatin repair is heterogeneous in the genome and is affected by multiple factors including transcription and chromatin states. Thus, the overall effect of damages in the genome is primarily driven not by damage formation but by the repair efficiency. The combination of the Damage-seq and XR-seq methods has the potential for developing novel cancer therapeutic strategies.


Assuntos
Cisplatino/farmacologia , Dano ao DNA/genética , Genoma Humano , Nucleotídeos/genética , Sequência de Bases , Linhagem Celular , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Humanos , Nucleossomos/metabolismo
11.
Genome Res ; 25(11): 1715-26, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26335634

RESUMO

The transition from a specified germ cell to a population of pluripotent cells occurs rapidly following fertilization. During this developmental transition, the zygotic genome is largely transcriptionally quiescent and undergoes significant chromatin remodeling. In Drosophila, the DNA-binding protein Zelda (also known as Vielfaltig) is required for this transition and for transcriptional activation of the zygotic genome. Open chromatin is associated with Zelda-bound loci, as well as more generally with regions of active transcription. Nonetheless, the extent to which Zelda influences chromatin accessibility across the genome is largely unknown. Here we used formaldehyde-assisted isolation of regulatory elements to determine the role of Zelda in regulating regions of open chromatin in the early embryo. We demonstrate that Zelda is essential for hundreds of regions of open chromatin. This Zelda-mediated chromatin accessibility facilitates transcription-factor recruitment and early gene expression. Thus, Zelda possesses some key characteristics of a pioneer factor. Unexpectedly, chromatin at a large subset of Zelda-bound regions remains open even in the absence of Zelda. The GAGA factor-binding motif and embryonic GAGA factor binding are specifically enriched in these regions. We propose that both Zelda and GAGA factor function to specify sites of open chromatin and together facilitate the remodeling of the early embryonic genome.


Assuntos
Cromatina/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/metabolismo , Animais , Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Feminino , Estudos de Associação Genética , Loci Gênicos , Masculino , Proteínas Nucleares , Regiões Promotoras Genéticas , Sequências Reguladoras de Ácido Nucleico , Análise de Sequência de DNA , Fatores de Transcrição/genética , Ativação Transcricional
12.
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
13.
Nature ; 484(7393): 251-5, 2012 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22498630

RESUMO

Dynamic access to genetic information is central to organismal development and environmental response. Consequently, genomic processes must be regulated by mechanisms that alter genome function relatively rapidly. Conventional chromatin immunoprecipitation (ChIP) experiments measure transcription factor occupancy, but give no indication of kinetics and are poor predictors of transcription factor function at a given locus. To measure transcription-factor-binding dynamics across the genome, we performed competition ChIP (refs 6, 7) with a sequence-specific Saccharomyces cerevisiae transcription factor, Rap1 (ref. 8). Rap1-binding dynamics and Rap1 occupancy were only weakly correlated (R(2) = 0.14), but binding dynamics were more strongly linked to function than occupancy. Long Rap1 residence was coupled to transcriptional activation, whereas fast binding turnover, which we refer to as 'treadmilling', was linked to low transcriptional output. Thus, DNA-binding events that seem identical by conventional ChIP may have different underlying modes of interaction that lead to opposing functional outcomes. We propose that transcription factor binding turnover is a major point of regulation in determining the functional consequences of transcription factor binding, and is mediated mainly by control of competition between transcription factors and nucleosomes. Our model predicts a clutch-like mechanism that rapidly engages a treadmilling transcription factor into a stable binding state, or vice versa, to modulate transcription factor function.


Assuntos
DNA Fúngico/metabolismo , Genoma Fúngico , Modelos Biológicos , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Bases , Sítios de Ligação , Ligação Competitiva , Imunoprecipitação da Cromatina , DNA Fúngico/genética , Regulação Fúngica da Expressão Gênica , Histona Acetiltransferases/metabolismo , Nucleossomos/genética , Nucleossomos/metabolismo , Ligação Proteica , RNA Polimerase II/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Saccharomyces cerevisiae/classificação , Complexo Shelterina , Fatores de Tempo
14.
Nature ; 484(7395): 534-7, 2012 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-22495302

RESUMO

Centromeres are chromosomal loci that direct segregation of the genome during cell division. The histone H3 variant CENP-A (also known as CenH3) defines centromeres in monocentric organisms, which confine centromere activity to a discrete chromosomal region, and holocentric organisms, which distribute centromere activity along the chromosome length. Because the highly repetitive DNA found at most centromeres is neither necessary nor sufficient for centromere function, stable inheritance of CENP-A nucleosomal chromatin is postulated to propagate centromere identity epigenetically. Here, we show that in the holocentric nematode Caenorhabditis elegans pre-existing CENP-A nucleosomes are not necessary to guide recruitment of new CENP-A nucleosomes. This is indicated by lack of CENP-A transmission by sperm during fertilization and by removal and subsequent reloading of CENP-A during oogenic meiotic prophase. Genome-wide mapping of CENP-A location in embryos and quantification of CENP-A molecules in nuclei revealed that CENP-A is incorporated at low density in domains that cumulatively encompass half the genome. Embryonic CENP-A domains are established in a pattern inverse to regions that are transcribed in the germline and early embryo, and ectopic transcription of genes in a mutant germline altered the pattern of CENP-A incorporation in embryos. Furthermore, regions transcribed in the germline but not embryos fail to incorporate CENP-A throughout embryogenesis. We propose that germline transcription defines genomic regions that exclude CENP-A incorporation in progeny, and that zygotic transcription during early embryogenesis remodels and reinforces this basal pattern. These findings link centromere identity to transcription and shed light on the evolutionary plasticity of centromeres.


Assuntos
Caenorhabditis elegans/genética , Centrômero/genética , Cromatina/genética , Células Germinativas/metabolismo , Transcrição Gênica , Animais , Autoantígenos/metabolismo , Evolução Biológica , Caenorhabditis elegans/embriologia , Proteína Centromérica A , Proteínas Cromossômicas não Histona/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/genética , Feminino , Fertilização , Regulação da Expressão Gênica no Desenvolvimento , Genoma Helmíntico , Gônadas/citologia , Gônadas/metabolismo , Organismos Hermafroditas , Masculino , Meiose
15.
PLoS Genet ; 11(4): e1005117, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25875092

RESUMO

After fertilization but prior to the onset of zygotic transcription, the C. elegans zygote cleaves asymmetrically to create the anterior AB and posterior P1 blastomeres, each of which goes on to generate distinct cell lineages. To understand how patterns of RNA inheritance and abundance arise after this first asymmetric cell division, we pooled hand-dissected AB and P1 blastomeres and performed RNA-seq. Our approach identified over 200 asymmetrically abundant mRNA transcripts. We confirmed symmetric or asymmetric abundance patterns for a subset of these transcripts using smFISH. smFISH also revealed heterogeneous subcellular patterning of the P1-enriched transcripts chs-1 and bpl-1. We screened transcripts enriched in a given blastomere for embryonic defects using RNAi. The gene neg-1 (F32D1.6) encoded an AB-enriched (anterior) transcript and was required for proper morphology of anterior tissues. In addition, analysis of the asymmetric transcripts yielded clues regarding the post-transcriptional mechanisms that control cellular mRNA abundance during asymmetric cell divisions, which are common in developing organisms.


Assuntos
Divisão Celular Assimétrica , Blastômeros/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Morfogênese , Proteínas Nucleares/metabolismo , RNA Mensageiro/metabolismo , Animais , Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas Nucleares/genética , RNA Mensageiro/genética
17.
Eur J Immunol ; 46(8): 1912-25, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27159132

RESUMO

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.


Assuntos
Cromatina/metabolismo , Inflamação/imunologia , Interleucina-10/genética , Intestinos/fisiopatologia , Macrófagos/metabolismo , Animais , Citocinas/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Expressão Gênica , Humanos , Tolerância Imunológica , Intestinos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
18.
Genome Res ; 24(2): 241-50, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24158655

RESUMO

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.


Assuntos
Carcinoma de Células Renais/genética , Cromatina/genética , Histona-Lisina N-Metiltransferase/genética , Neoplasias Renais/genética , Carcinoma de Células Renais/patologia , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Neoplasias Renais/patologia , Mutação , Proteínas Nucleares/genética , Processamento Pós-Transcricional do RNA/genética , Splicing de RNA/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética
19.
Mol Cell ; 34(6): 722-34, 2009 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-19560424

RESUMO

Replication forks face multiple obstacles that slow their progression. By two-dimensional gel analysis, yeast forks pause at stable DNA protein complexes, and this pausing is greatly increased in the absence of the Rrm3 helicase. We used a genome-wide approach to identify 96 sites of very high DNA polymerase binding in wild-type cells. Most of these binding sites were not previously identified pause sites. Rather, the most highly represented genomic category among high DNA polymerase binding sites was the open reading frames (ORFs) of highly transcribed RNA polymerase II genes. Twice as many pause sites were identified in rrm3 compared with wild-type cells, as pausing in this strain occurred at both highly transcribed RNA polymerase II genes and the previously identified protein DNA complexes. ORFs of highly transcribed RNA polymerase II genes are a class of natural pause sites that are not exacerbated in rrm3 cells.


Assuntos
DNA Helicases/metabolismo , DNA Polimerase II/metabolismo , Replicação do DNA/fisiologia , RNA Polimerase II/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Transcrição Gênica , Sítios de Ligação , DNA Helicases/genética , Mutação , Fases de Leitura Aberta , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Complexo Shelterina , Proteínas de Ligação a Telômeros/metabolismo , Fatores de Transcrição/metabolismo
20.
PLoS Genet ; 10(11): e1004798, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25411781

RESUMO

Associating genetic variation with quantitative measures of gene regulation offers a way to bridge the gap between genotype and complex phenotypes. In order to identify quantitative trait loci (QTLs) that influence the binding of a transcription factor in humans, we measured binding of the multifunctional transcription and chromatin factor CTCF in 51 HapMap cell lines. We identified thousands of QTLs in which genotype differences were associated with differences in CTCF binding strength, hundreds of them confirmed by directly observable allele-specific binding bias. The majority of QTLs were either within 1 kb of the CTCF binding motif, or in linkage disequilibrium with a variant within 1 kb of the motif. On the X chromosome we observed three classes of binding sites: a minority class bound only to the active copy of the X chromosome, the majority class bound to both the active and inactive X, and a small set of female-specific CTCF sites associated with two non-coding RNA genes. In sum, our data reveal extensive genetic effects on CTCF binding, both direct and indirect, and identify a diversity of patterns of CTCF binding on the X chromosome.


Assuntos
Cromossomos Humanos X/genética , Locos de Características Quantitativas , Proteínas Repressoras/genética , Alelos , Fator de Ligação a CCCTC , Feminino , Humanos , Desequilíbrio de Ligação , Ligação Proteica , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Proteínas Repressoras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA