Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Cell ; 163(6): 1375-87, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26638071

RESUMEN

TFIIB-related factor 2 (Brf2) is a member of the family of TFIIB-like core transcription factors. Brf2 recruits RNA polymerase (Pol) III to type III gene-external promoters, including the U6 spliceosomal RNA and selenocysteine tRNA genes. Found only in vertebrates, Brf2 has been linked to tumorigenesis but the underlying mechanisms remain elusive. We have solved crystal structures of a human Brf2-TBP complex bound to natural promoters, obtaining a detailed view of the molecular interactions occurring at Brf2-dependent Pol III promoters and highlighting the general structural and functional conservation of human Pol II and Pol III pre-initiation complexes. Surprisingly, our structural and functional studies unravel a Brf2 redox-sensing module capable of specifically regulating Pol III transcriptional output in living cells. Furthermore, we establish Brf2 as a central redox-sensing transcription factor involved in the oxidative stress pathway and provide a mechanistic model for Brf2 genetic activation in lung and breast cancer.


Asunto(s)
Oxidación-Reducción , Factor de Transcripción TFIIIB/química , Factor de Transcripción TFIIIB/metabolismo , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , ADN/química , ADN/metabolismo , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , ARN Polimerasa III/metabolismo , Saccharomyces cerevisiae , Alineación de Secuencia , Transducción de Señal
2.
Cell ; 162(5): 1039-50, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26300124

RESUMEN

Chromatin state variation at gene regulatory elements is abundant across individuals, yet we understand little about the genetic basis of this variability. Here, we profiled several histone modifications, the transcription factor (TF) PU.1, RNA polymerase II, and gene expression in lymphoblastoid cell lines from 47 whole-genome sequenced individuals. We observed that distinct cis-regulatory elements exhibit coordinated chromatin variation across individuals in the form of variable chromatin modules (VCMs) at sub-Mb scale. VCMs were associated with thousands of genes and preferentially cluster within chromosomal contact domains. We mapped strong proximal and weak, yet more ubiquitous, distal-acting chromatin quantitative trait loci (cQTL) that frequently explain this variation. cQTLs were associated with molecular activity at clusters of cis-regulatory elements and mapped preferentially within TF-bound regions. We propose that local, sequence-independent chromatin variation emerges as a result of genetic perturbations in cooperative interactions between cis-regulatory elements that are located within the same genomic domain.


Asunto(s)
Cromatina/química , Regulación de la Expresión Génica , Variación Genética , Genoma Humano , Cromatina/metabolismo , Cromosomas Humanos/química , Genética de Población , Humanos , Sitios de Carácter Cuantitativo , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/metabolismo
3.
Genes Dev ; 34(13-14): 865-882, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32611613

RESUMEN

RNA polymerase (Pol) III is responsible for transcription of different noncoding genes in eukaryotic cells, whose RNA products have well-defined functions in translation and other biological processes for some, and functions that remain to be defined for others. For all of them, however, new functions are being described. For example, Pol III products have been reported to regulate certain proteins such as protein kinase R (PKR) by direct association, to constitute the source of very short RNAs with regulatory roles in gene expression, or to control microRNA levels by sequestration. Consistent with these many functions, deregulation of Pol III transcribed genes is associated with a large variety of human disorders. Here we review different human diseases that have been linked to defects in the Pol III transcription apparatus or to Pol III products imbalance and discuss the possible underlying mechanisms.


Asunto(s)
Enfermedad/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Neoplasias/genética , ARN Polimerasa III/genética , ARN Polimerasa III/metabolismo , Transcripción Genética , Animales , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/enzimología , Humanos , Mutación , Neoplasias/enzimología , ARN de Transferencia/genética , ARN de Transferencia/metabolismo
4.
Genes Dev ; 32(9-10): 711-722, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29785964

RESUMEN

RNA polymerase II (Pol II) small nuclear RNA (snRNA) promoters and type 3 Pol III promoters have highly similar structures; both contain an interchangeable enhancer and "proximal sequence element" (PSE), which recruits the SNAP complex (SNAPc). The main distinguishing feature is the presence, in the type 3 promoters only, of a TATA box, which determines Pol III specificity. To understand the mechanism by which the absence or presence of a TATA box results in specific Pol recruitment, we examined how SNAPc and general transcription factors required for Pol II or Pol III transcription of SNAPc-dependent genes (i.e., TATA-box-binding protein [TBP], TFIIB, and TFIIA for Pol II transcription and TBP and BRF2 for Pol III transcription) assemble to ensure specific Pol recruitment. TFIIB and BRF2 could each, in a mutually exclusive fashion, be recruited to SNAPc. In contrast, TBP-TFIIB and TBP-BRF2 complexes were not recruited unless a TATA box was present, which allowed selective and efficient recruitment of the TBP-BRF2 complex. Thus, TBP both prevented BRF2 recruitment to Pol II promoters and enhanced BRF2 recruitment to Pol III promoters. On Pol II promoters, TBP recruitment was separate from TFIIB recruitment and enhanced by TFIIA. Our results provide a model for specific Pol recruitment at SNAPc-dependent promoters.


Asunto(s)
Regiones Promotoras Genéticas , ARN Polimerasa III/metabolismo , ARN Polimerasa II/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Células HEK293 , Humanos , Mutación , Unión Proteica , Dominios Proteicos , Transporte de Proteínas , TATA Box/genética , Proteína de Unión a TATA-Box/metabolismo , Factor de Transcripción TFIIB/metabolismo , Factores de Transcripción/metabolismo
5.
Genes Dev ; 31(4): 413-421, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28289142

RESUMEN

Overlapping gene arrangements can potentially contribute to gene expression regulation. A mammalian interspersed repeat (MIR) nested in antisense orientation within the first intron of the Polr3e gene, encoding an RNA polymerase III (Pol III) subunit, is conserved in mammals and highly occupied by Pol III. Using a fluorescence assay, CRISPR/Cas9-mediated deletion of the MIR in mouse embryonic stem cells, and chromatin immunoprecipitation assays, we show that the MIR affects Polr3e expression through transcriptional interference. Our study reveals a mechanism by which a Pol II gene can be regulated at the transcription elongation level by transcription of an embedded antisense Pol III gene.


Asunto(s)
Regulación de la Expresión Génica/genética , ARN Polimerasa III/metabolismo , Animales , Secuencia Conservada/genética , ADN sin Sentido/genética , Células Madre Embrionarias , Secuencias Repetitivas Esparcidas/genética , Intrones/genética , Ratones , ARN Polimerasa II/genética , ARN Polimerasa III/genética , ARN Mensajero/genética , Eliminación de Secuencia
6.
Genes Dev ; 29(9): 934-47, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25934505

RESUMEN

MAF1 is a global repressor of RNA polymerase III transcription that regulates the expression of highly abundant noncoding RNAs in response to nutrient availability and cellular stress. Thus, MAF1 function is thought to be important for metabolic economy. Here we show that a whole-body knockout of Maf1 in mice confers resistance to diet-induced obesity and nonalcoholic fatty liver disease by reducing food intake and increasing metabolic inefficiency. Energy expenditure in Maf1(-/-) mice is increased by several mechanisms. Precursor tRNA synthesis was increased in multiple tissues without significant effects on mature tRNA levels, implying increased turnover in a futile tRNA cycle. Elevated futile cycling of hepatic lipids was also observed. Metabolite profiling of the liver and skeletal muscle revealed elevated levels of many amino acids and spermidine, which links the induction of autophagy in Maf1(-/-) mice with their extended life span. The increase in spermidine was accompanied by reduced levels of nicotinamide N-methyltransferase, which promotes polyamine synthesis, enables nicotinamide salvage to regenerate NAD(+), and is associated with obesity resistance. Consistent with this, NAD(+) levels were increased in muscle. The importance of MAF1 for metabolic economy reveals the potential for MAF1 modulators to protect against obesity and its harmful consequences.


Asunto(s)
Proteínas Represoras/genética , Animales , Autofagia/genética , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Metabolismo de los Lípidos/genética , Longevidad/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/genética , ARN de Transferencia/metabolismo , Espermidina/metabolismo
7.
Trends Genet ; 35(6): 457-469, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31040056

RESUMEN

Nuclear eukaryotic genomes are transcribed by three related RNA polymerases (Pol), which transcribe distinct gene sets. Specific Pol recruitment is achieved through selective core promoter recognition by basal transcription factors (TFs). Transcription by an inappropriate Pol appears to be rare and to generate mostly unstable products. A collection of short noncoding RNA genes [for example, small nuclear RNA (snRNA) or 7SK RNA genes], which play essential roles in processes such as maturation of RNA molecules or control of Pol II transcription elongation, possess highly similar core promoters, and yet are transcribed for some by Pol II and for others by Pol III as a result of small promoter differences. Here we discuss the mechanisms of selective Pol recruitment to such promoters.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación de la Expresión Génica , ARN Nuclear Pequeño/genética , Animales , Biomarcadores , ARN Polimerasas Dirigidas por ADN/genética , Humanos , Complejos Multiproteicos/metabolismo , Especificidad de Órganos/genética , Regiones Promotoras Genéticas , Unión Proteica , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , ARN Nuclear Pequeño/metabolismo , Transcripción Genética
8.
Nucleic Acids Res ; 47(4): 1786-1796, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30597109

RESUMEN

Mouse liver regeneration after partial hepatectomy involves cells in the remaining tissue synchronously entering the cell division cycle. We have used this system and H3K4me3, Pol II and Pol III profiling to characterize adaptations in Pol III transcription. Our results broadly define a class of genes close to H3K4me3 and Pol II peaks, whose Pol III occupancy is high and stable, and another class, distant from Pol II peaks, whose Pol III occupancy strongly increases after partial hepatectomy. Pol III regulation in the liver thus entails both highly expressed housekeeping genes and genes whose expression can adapt to increased demand.


Asunto(s)
Regeneración Hepática/genética , Hígado/crecimiento & desarrollo , ARN Polimerasa III/genética , Transcripción Genética , Animales , Ciclo Celular/genética , División Celular/genética , Inmunoprecipitación de Cromatina , Regulación del Desarrollo de la Expresión Génica/genética , Hepatectomía , N-Metiltransferasa de Histona-Lisina/química , N-Metiltransferasa de Histona-Lisina/genética , Histonas/química , Histonas/genética , Humanos , Hígado/patología , Hígado/cirugía , Ratones , Unión Proteica , ARN Polimerasa II/química , ARN Polimerasa II/genética , ARN Polimerasa III/química
9.
Proc Natl Acad Sci U S A ; 115(48): 12182-12187, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30429315

RESUMEN

As a master negative regulator of RNA polymerase (Pol) III, Maf1 modulates transcription in response to nutrients and stress to balance the production of highly abundant tRNAs, 5S rRNA, and other small noncoding RNAs with cell growth and maintenance. This regulation of Pol III transcription is important for energetic economy as mice lacking Maf1 are lean and resist weight gain on normal and high fat diets. The lean phenotype of Maf1 knockout (KO) mice is attributed in part to metabolic inefficiencies which increase the demand for cellular energy and elevate catabolic processes, including autophagy/lipophagy and lipolysis. A futile RNA cycle involving increased synthesis and turnover of Pol III transcripts has been proposed as an important driver of these changes. Here, using targeted metabolomics, we find changes in the liver of fed and fasted Maf1 KO mice consistent with the function of mammalian Maf1 as a chronic Pol III repressor. Differences in long-chain acylcarnitine levels suggest that energy demand is higher in the fed state of Maf1 KO mice versus the fasted state. Quantitative metabolite profiling supports increased activity in the TCA cycle, the pentose phosphate pathway, and the urea cycle and reveals changes in nucleotide levels and the creatine system. Metabolite profiling also confirms key predictions of the futile RNA cycle hypothesis by identifying changes in many metabolites involved in nucleotide synthesis and turnover. Thus, constitutively high levels of Pol III transcription in Maf1 KO mice reprogram central metabolic pathways and waste metabolic energy through a futile RNA cycle.


Asunto(s)
Peso Corporal , Metaboloma , ARN Polimerasa III/metabolismo , Animales , Regulación de la Expresión Génica , Hígado/metabolismo , Redes y Vías Metabólicas , Ratones , Ratones Noqueados , Fenotipo , ARN Polimerasa III/genética , ARN Ribosómico 5S/genética , ARN Ribosómico 5S/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
10.
Genome Res ; 27(6): 973-984, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28341772

RESUMEN

RNA polymerase III (Pol III) synthesizes short noncoding RNAs, many of which are essential for translation. Accordingly, Pol III activity is tightly regulated with cell growth and proliferation by factors such as MYC, RB1, TRP53, and MAF1. MAF1 is a repressor of Pol III transcription whose activity is controlled by phosphorylation; in particular, it is inactivated through phosphorylation by the TORC1 kinase complex, a sensor of nutrient availability. Pol III regulation is thus sensitive to environmental cues, yet a diurnal profile of Pol III transcription activity is so far lacking. Here, we first use gene expression arrays to measure mRNA accumulation during the diurnal cycle in the livers of (1) wild-type mice, (2) arrhythmic Arntl knockout mice, (3) mice fed at regular intervals during both night and day, and (4) mice lacking the Maf1 gene, and so provide a comprehensive view of the changes in cyclic mRNA accumulation occurring in these different systems. We then show that Pol III occupancy of its target genes rises before the onset of the night, stays high during the night, when mice normally ingest food and when translation is known to be increased, and decreases in daytime. Whereas higher Pol III occupancy during the night reflects a MAF1-dependent response to feeding, the rise of Pol III occupancy before the onset of the night reflects a circadian clock-dependent response. Thus, Pol III transcription during the diurnal cycle is regulated both in response to nutrients and by the circadian clock, which allows anticipatory Pol III transcription.


Asunto(s)
Factores de Transcripción ARNTL/genética , Relojes Circadianos/genética , Ritmo Circadiano/genética , ARN Polimerasa III/genética , Proteínas Represoras/genética , Transcripción Genética , Factores de Transcripción ARNTL/deficiencia , Animales , Ingestión de Alimentos/genética , Ayuno/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hígado/metabolismo , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , ARN Polimerasa III/metabolismo , Proteínas Represoras/deficiencia , Transducción de Señal
12.
PLoS Biol ; 15(4): e2001069, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28414715

RESUMEN

Many organisms exhibit temporal rhythms in gene expression that propel diurnal cycles in physiology. In the liver of mammals, these rhythms are controlled by transcription-translation feedback loops of the core circadian clock and by feeding-fasting cycles. To better understand the regulatory interplay between the circadian clock and feeding rhythms, we mapped DNase I hypersensitive sites (DHSs) in the mouse liver during a diurnal cycle. The intensity of DNase I cleavages cycled at a substantial fraction of all DHSs, suggesting that DHSs harbor regulatory elements that control rhythmic transcription. Using chromatin immunoprecipitation followed by DNA sequencing (ChIP-seq), we found that hypersensitivity cycled in phase with RNA polymerase II (Pol II) loading and H3K27ac histone marks. We then combined the DHSs with temporal Pol II profiles in wild-type (WT) and Bmal1-/- livers to computationally identify transcription factors through which the core clock and feeding-fasting cycles control diurnal rhythms in transcription. While a similar number of mRNAs accumulated rhythmically in Bmal1-/- compared to WT livers, the amplitudes in Bmal1-/- were generally lower. The residual rhythms in Bmal1-/- reflected transcriptional regulators mediating feeding-fasting responses as well as responses to rhythmic systemic signals. Finally, the analysis of DNase I cuts at nucleotide resolution showed dynamically changing footprints consistent with dynamic binding of CLOCK:BMAL1 complexes. Structural modeling suggested that these footprints are driven by a transient heterotetramer binding configuration at peak activity. Together, our temporal DNase I mappings allowed us to decipher the global regulation of diurnal transcription rhythms in the mouse liver.


Asunto(s)
Ritmo Circadiano/genética , Regulación de la Expresión Génica , Hígado/fisiología , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Inmunoprecipitación de Cromatina , Relojes Circadianos/genética , Desoxirribonucleasa I/genética , Desoxirribonucleasa I/metabolismo , Ayuno , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , Factores de Transcripción/genética , Transcripción Genética
13.
Genes Dev ; 26(15): 1643-7, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22855826

RESUMEN

In this issue of Genes & Development, Revyakin and colleagues (pp. 1691-1702) measure the relation between individual RNA polymerase II transcription events and transcription factor assembly by counting RNA transcripts retained on the template DNA using single-molecule fluorescence.


Asunto(s)
Imagen Molecular/métodos , ARN Polimerasa II/química , Transcripción Genética , Humanos
14.
Genome Res ; 26(5): 624-35, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26941251

RESUMEN

RNA polymerase III (Pol III) is tightly controlled in response to environmental cues, yet a genomic-scale picture of Pol III regulation and the role played by its repressor MAF1 is lacking. Here, we describe genome-wide studies in human fibroblasts that reveal a dynamic and gene-specific adaptation of Pol III recruitment to extracellular signals in an mTORC1-dependent manner. Repression of Pol III recruitment and transcription are tightly linked to MAF1, which selectively localizes at Pol III loci, even under serum-replete conditions, and increasingly targets transcribing Pol III in response to serum starvation. Combining Pol III binding profiles with EU-labeling and high-throughput sequencing of newly synthesized small RNAs, we show that Pol III occupancy closely reflects ongoing transcription. Our results exclude the long-term, unproductive arrest of Pol III on the DNA as a major regulatory mechanism and identify previously uncharacterized, differential coordination in Pol III binding and transcription under different growth conditions.


Asunto(s)
Complejos Multiproteicos/metabolismo , ARN Polimerasa III/metabolismo , Proteínas Represoras/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transcripción Genética/fisiología , Línea Celular , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , ARN Polimerasa III/genética , Proteínas Represoras/genética , Serina-Treonina Quinasas TOR/genética
15.
Genome Res ; 24(1): 37-51, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24107381

RESUMEN

RNA polymerase III (Pol III) occurs in two versions, one containing the POLR3G subunit and the other the closely related POLR3GL subunit. It is not clear whether these two Pol III forms have the same function, in particular whether they recognize the same target genes. We show that the POLR3G and POLR3GL genes arose from a DNA-based gene duplication, probably in a common ancestor of vertebrates. POLR3G- as well as POLR3GL-containing Pol III are present in cultured cell lines and in normal mouse liver, although the relative amounts of the two forms vary, with the POLR3G-containing Pol III relatively more abundant in dividing cells. Genome-wide chromatin immunoprecipitations followed by high-throughput sequencing (ChIP-seq) reveal that both forms of Pol III occupy the same target genes, in very constant proportions within one cell line, suggesting that the two forms of Pol III have a similar function with regard to specificity for target genes. In contrast, the POLR3G promoter--not the POLR3GL promoter--binds the transcription factor MYC, as do all other promoters of genes encoding Pol III subunits. Thus, the POLR3G/POLR3GL duplication did not lead to neo-functionalization of the gene product (at least with regard to target gene specificity) but rather to neo-functionalization of the transcription units, which acquired different mechanisms of regulation, thus likely affording greater regulation potential to the cell.


Asunto(s)
Duplicación de Gen , ARN Polimerasa III/genética , ARN Polimerasa III/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Cromatina/metabolismo , Evolución Molecular , Genoma , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Filogenia , Regiones Promotoras Genéticas , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Polimerasa III/química , Alineación de Secuencia , Vertebrados/genética
16.
Genome Res ; 24(7): 1157-68, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24709819

RESUMEN

Chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) experiments are widely used to determine, within entire genomes, the occupancy sites of any protein of interest, including, for example, transcription factors, RNA polymerases, or histones with or without various modifications. In addition to allowing the determination of occupancy sites within one cell type and under one condition, this method allows, in principle, the establishment and comparison of occupancy maps in various cell types, tissues, and conditions. Such comparisons require, however, that samples be normalized. Widely used normalization methods that include a quantile normalization step perform well when factor occupancy varies at a subset of sites, but may miss uniform genome-wide increases or decreases in site occupancy. We describe a spike adjustment procedure (SAP) that, unlike commonly used normalization methods intervening at the analysis stage, entails an experimental step prior to immunoprecipitation. A constant, low amount from a single batch of chromatin of a foreign genome is added to the experimental chromatin. This "spike" chromatin then serves as an internal control to which the experimental signals can be adjusted. We show that the method improves similarity between replicates and reveals biological differences including global and largely uniform changes.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Animales , Cromatina/genética , Cromatina/metabolismo , Inmunoprecipitación de Cromatina/normas , Biología Computacional/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Humanos , Ratones , Control de Calidad , Estándares de Referencia , Reproducibilidad de los Resultados
17.
Genome Res ; 22(4): 666-80, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22287103

RESUMEN

The genomic loci occupied by RNA polymerase (RNAP) III have been characterized in human culture cells by genome-wide chromatin immunoprecipitations, followed by deep sequencing (ChIP-seq). These studies have shown that only ∼40% of the annotated 622 human tRNA genes and pseudogenes are occupied by RNAP-III, and that these genes are often in open chromatin regions rich in active RNAP-II transcription units. We have used ChIP-seq to characterize RNAP-III-occupied loci in a differentiated tissue, the mouse liver. Our studies define the mouse liver RNAP-III-occupied loci including a conserved mammalian interspersed repeat (MIR) as a potential regulator of an RNAP-III subunit-encoding gene. They reveal that synteny relationships can be established between a number of human and mouse RNAP-III genes, and that the expression levels of these genes are significantly linked. They establish that variations within the A and B promoter boxes, as well as the strength of the terminator sequence, can strongly affect RNAP-III occupancy of tRNA genes. They reveal correlations with various genomic features that explain the observed variation of 81% of tRNA scores. In mouse liver, loci represented in the NCBI37/mm9 genome assembly that are clearly occupied by RNAP-III comprise 50 Rn5s (5S RNA) genes, 14 known non-tRNA RNAP-III genes, nine Rn4.5s (4.5S RNA) genes, and 29 SINEs. Moreover, out of the 433 annotated tRNA genes, half are occupied by RNAP-III. Transfer RNA gene expression levels reflect both an underlying genomic organization conserved in dividing human culture cells and resting mouse liver cells, and the particular promoter and terminator strengths of individual genes.


Asunto(s)
Perfilación de la Expresión Génica , Genómica/métodos , Hígado/metabolismo , ARN Polimerasa III/genética , Animales , Inmunoprecipitación de Cromatina/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Polimerasa III/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Análisis de Secuencia de ADN/métodos
18.
Bioinformatics ; 30(2): 165-71, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24255646

RESUMEN

MOTIVATION: High-throughput sequencing technologies enable the genome-wide analysis of the impact of genetic variation on molecular phenotypes at unprecedented resolution. However, although powerful, these technologies can also introduce unexpected artifacts. RESULTS: We investigated the impact of library amplification bias on the identification of allele-specific (AS) molecular events from high-throughput sequencing data derived from chromatin immunoprecipitation assays (ChIP-seq). Putative AS DNA binding activity for RNA polymerase II was determined using ChIP-seq data derived from lymphoblastoid cell lines of two parent-daughter trios. We found that, at high-sequencing depth, many significant AS binding sites suffered from an amplification bias, as evidenced by a larger number of clonal reads representing one of the two alleles. To alleviate this bias, we devised an amplification bias detection strategy, which filters out sites with low read complexity and sites featuring a significant excess of clonal reads. This method will be useful for AS analyses involving ChIP-seq and other functional sequencing assays. AVAILABILITY: The R package abs filter for library clonality simulations and detection of amplification-biased sites is available from http://updepla1srv1.epfl.ch/waszaks/absfilter


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Genoma Humano , Linfocitos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Polimerasa II/genética , Alelos , Sitios de Unión , Femenino , Biblioteca de Genes , Humanos , Linfocitos/citología , Masculino , Polimorfismo de Nucleótido Simple/genética
19.
PLoS Biol ; 10(11): e1001442, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209382

RESUMEN

Interactions of cell-autonomous circadian oscillators with diurnal cycles govern the temporal compartmentalization of cell physiology in mammals. To understand the transcriptional and epigenetic basis of diurnal rhythms in mouse liver genome-wide, we generated temporal DNA occupancy profiles by RNA polymerase II (Pol II) as well as profiles of the histone modifications H3K4me3 and H3K36me3. We used these data to quantify the relationships of phases and amplitudes between different marks. We found that rhythmic Pol II recruitment at promoters rather than rhythmic transition from paused to productive elongation underlies diurnal gene transcription, a conclusion further supported by modeling. Moreover, Pol II occupancy preceded mRNA accumulation by 3 hours, consistent with mRNA half-lives. Both methylation marks showed that the epigenetic landscape is highly dynamic and globally remodeled during the 24-hour cycle. While promoters of transcribed genes had tri-methylated H3K4 even at their trough activity times, tri-methylation levels reached their peak, on average, 1 hour after Pol II. Meanwhile, rhythms in tri-methylation of H3K36 lagged transcription by 3 hours. Finally, modeling profiles of Pol II occupancy and mRNA accumulation identified three classes of genes: one showing rhythmicity both in transcriptional and mRNA accumulation, a second class with rhythmic transcription but flat mRNA levels, and a third with constant transcription but rhythmic mRNAs. The latter class emphasizes widespread temporally gated posttranscriptional regulation in the mouse liver.


Asunto(s)
Ritmo Circadiano , Epigénesis Genética , ARN Polimerasa II/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética , Animales , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Metilación de ADN , Semivida , Histonas/genética , Histonas/metabolismo , Cinética , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Sitio de Iniciación de la Transcripción , Transcriptoma
20.
PLoS Genet ; 8(11): e1003028, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23166507

RESUMEN

SNAP(c) is one of a few basal transcription factors used by both RNA polymerase (pol) II and pol III. To define the set of active SNAP(c)-dependent promoters in human cells, we have localized genome-wide four SNAP(c) subunits, GTF2B (TFIIB), BRF2, pol II, and pol III. Among some seventy loci occupied by SNAP(c) and other factors, including pol II snRNA genes, pol III genes with type 3 promoters, and a few un-annotated loci, most are primarily occupied by either pol II and GTF2B, or pol III and BRF2. A notable exception is the RPPH1 gene, which is occupied by significant amounts of both polymerases. We show that the large majority of SNAP(c)-dependent promoters recruit POU2F1 and/or ZNF143 on their enhancer region, and a subset also recruits GABP, a factor newly implicated in SNAP(c)-dependent transcription. These activators associate with pol II and III promoters in G1 slightly before the polymerase, and ZNF143 is required for efficient transcription initiation complex assembly. The results characterize a set of genes with unique properties and establish that polymerase specificity is not absolute in vivo.


Asunto(s)
Regiones Promotoras Genéticas , ARN Polimerasa III , ARN Polimerasa II , Factores de Transcripción , Proteínas de Unión al ADN/genética , Genoma Humano , Humanos , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa III/genética , ARN Polimerasa III/metabolismo , ARN Nuclear Pequeño/genética , Secuencias Reguladoras de Ácidos Nucleicos , Factor de Transcripción TFIIB/genética , Factor de Transcripción TFIIB/metabolismo , Factor de Transcripción TFIIIB/genética , Factor de Transcripción TFIIIB/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA