Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Science ; 376(6588): eabk3112, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35357925

RESUMEN

Mobile elements and repetitive genomic regions are sources of lineage-specific genomic innovation and uniquely fingerprint individual genomes. Comprehensive analyses of such repeat elements, including those found in more complex regions of the genome, require a complete, linear genome assembly. We present a de novo repeat discovery and annotation of the T2T-CHM13 human reference genome. We identified previously unknown satellite arrays, expanded the catalog of variants and families for repeats and mobile elements, characterized classes of complex composite repeats, and located retroelement transduction events. We detected nascent transcription and delineated CpG methylation profiles to define the structure of transcriptionally active retroelements in humans, including those in centromeres. These data expand our insight into the diversity, distribution, and evolution of repetitive regions that have shaped the human genome.


Asunto(s)
Epigénesis Genética , Genoma Humano , Secuencias Repetitivas de Ácidos Nucleicos , Telómero/genética , Transcripción Genética , Humanos
2.
Mol Cell ; 81(8): 1732-1748.e8, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33730542

RESUMEN

During self-renewal, cell-type-defining features are drastically perturbed in mitosis and must be faithfully reestablished upon G1 entry, a process that remains largely elusive. Here, we characterized at a genome-wide scale the dynamic transcriptional and architectural resetting of mouse pluripotent stem cells (PSCs) upon mitotic exit. We captured distinct waves of transcriptional reactivation with rapid induction of stem cell genes and transient activation of lineage-specific genes. Topological reorganization at different hierarchical levels also occurred in an asynchronous manner and showed partial coordination with transcriptional resetting. Globally, rapid transcriptional and architectural resetting associated with mitotic retention of H3K27 acetylation, supporting a bookmarking function. Indeed, mitotic depletion of H3K27ac impaired the early reactivation of bookmarked, stem-cell-associated genes. However, 3D chromatin reorganization remained largely unaffected, suggesting that these processes are driven by distinct forces upon mitotic exit. This study uncovers principles and mediators of PSC molecular resetting during self-renewal.


Asunto(s)
Cromatina/genética , Código de Histonas/genética , Histonas/genética , Mitosis/genética , Células Madre Pluripotentes/fisiología , Acetilación , Animales , Línea Celular , Drosophila/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Transcripción Genética/genética , Activación Transcripcional/genética
3.
Genes Dev ; 33(19-20): 1441-1455, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31467088

RESUMEN

Rapid perturbation of protein function permits the ability to define primary molecular responses while avoiding downstream cumulative effects of protein dysregulation. The auxin-inducible degron (AID) system was developed as a tool to achieve rapid and inducible protein degradation in nonplant systems. However, tagging proteins at their endogenous loci results in chronic auxin-independent degradation by the proteasome. To correct this deficiency, we expressed the auxin response transcription factor (ARF) in an improved inducible degron system. ARF is absent from previously engineered AID systems but is a critical component of native auxin signaling. In plants, ARF directly interacts with AID in the absence of auxin, and we found that expression of the ARF PB1 (Phox and Bem1) domain suppresses constitutive degradation of AID-tagged proteins. Moreover, the rate of auxin-induced AID degradation is substantially faster in the ARF-AID system. To test the ARF-AID system in a quantitative and sensitive manner, we measured genome-wide changes in nascent transcription after rapidly depleting the ZNF143 transcription factor. Transcriptional profiling indicates that ZNF143 activates transcription in cis and regulates promoter-proximal paused RNA polymerase density. Rapidly inducible degradation systems that preserve the target protein's native expression levels and patterns will revolutionize the study of biological systems by enabling specific and temporally defined protein dysregulation.


Asunto(s)
Técnicas Genéticas , Proteínas/metabolismo , Proteolisis , Línea Celular , Inhibidores de Cisteína Proteinasa/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Ácidos Indolacéticos/farmacología , Leupeptinas/farmacología , Células MCF-7 , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos , Transactivadores/genética , Transactivadores/metabolismo
4.
Genes Dev ; 33(15-16): 960-982, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31123063

RESUMEN

Precise spatio-temporal control of gene activity is essential for organismal development, growth, and survival in a changing environment. Decisive steps in gene regulation involve the pausing of RNA polymerase II (Pol II) in early elongation, and the controlled release of paused polymerase into productive RNA synthesis. Here we describe the factors that enable pausing and the events that trigger Pol II release into the gene. We also discuss open questions in the field concerning the stability of paused Pol II, nucleosomes as obstacles to elongation, and potential roles of pausing in defining the precision and dynamics of gene expression.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , Animales , Estabilidad de Enzimas , Humanos , Nucleosomas/metabolismo , ARN Polimerasa II/genética , Elongación de la Transcripción Genética
5.
Proc Natl Acad Sci U S A ; 116(6): 2181-2186, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30674673

RESUMEN

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by the loss of function from the maternal allele of UBE3A, a gene encoding an E3 ubiquitin ligase. UBE3A is only expressed from the maternally inherited allele in mature human neurons due to tissue-specific genomic imprinting. Imprinted expression of UBE3A is restricted to neurons by expression of UBE3A antisense transcript (UBE3A-ATS) from the paternally inherited allele, which silences the paternal allele of UBE3A in cis However, the mechanism restricting UBE3A-ATS expression and UBE3A imprinting to neurons is not understood. We used CRISPR/Cas9-mediated genome editing to functionally define a bipartite boundary element critical for neuron-specific expression of UBE3A-ATS in humans. Removal of this element led to up-regulation of UBE3A-ATS without repressing paternal UBE3A However, increasing expression of UBE3A-ATS in the absence of the boundary element resulted in full repression of paternal UBE3A, demonstrating that UBE3A imprinting requires both the loss of function from the boundary element as well as the up-regulation of UBE3A-ATS These results suggest that manipulation of the competition between UBE3A-ATS and UBE3A may provide a potential therapeutic approach for AS.


Asunto(s)
Cromatina/genética , Impresión Genómica , Neuronas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Síndrome de Angelman/genética , Sitios de Unión , Cromatina/metabolismo , Epistasis Genética , Exones , Expresión Génica , Regulación de la Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Unión Proteica , ARN sin Sentido , ARN Largo no Codificante , Eliminación de Secuencia
6.
Genome Res ; 29(2): 223-235, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30606742

RESUMEN

The aberrant activities of transcription factors such as the androgen receptor (AR) underpin prostate cancer development. While the AR cis-regulation has been extensively studied in prostate cancer, information pertaining to the spatial architecture of the AR transcriptional circuitry remains limited. In this paper, we propose a novel framework to profile long-range chromatin interactions associated with AR and its collaborative transcription factor, erythroblast transformation-specific related gene (ERG), using chromatin interaction analysis by paired-end tag (ChIA-PET). We identified ERG-associated long-range chromatin interactions as a cooperative component in the AR-associated chromatin interactome, acting in concert to achieve coordinated regulation of a subset of AR target genes. Through multifaceted functional data analysis, we found that AR-ERG interaction hub regions are characterized by distinct functional signatures, including bidirectional transcription and cotranscription factor binding. In addition, cancer-associated long noncoding RNAs were found to be connected near protein-coding genes through AR-ERG looping. Finally, we found strong enrichment of prostate cancer genome-wide association study (GWAS) single nucleotide polymorphisms (SNPs) at AR-ERG co-binding sites participating in chromatin interactions and gene regulation, suggesting GWAS target genes identified from chromatin looping data provide more biologically relevant findings than using the nearest gene approach. Taken together, our results revealed an AR-ERG-centric higher-order chromatin structure that drives coordinated gene expression in prostate cancer progression and the identification of potential target genes for therapeutic intervention.


Asunto(s)
Cromatina/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/genética , Receptores Androgénicos/metabolismo , Transcripción Genética , Línea Celular Tumoral , Cromatina/química , Redes Reguladoras de Genes , Genoma Humano , Humanos , Masculino , Proteínas de Fusión Oncogénica/análisis , Polimorfismo de Nucleótido Simple , Neoplasias de la Próstata/metabolismo , ARN Largo no Codificante/metabolismo , Regulador Transcripcional ERG/metabolismo , Regulador Transcripcional ERG/fisiología
7.
Nat Genet ; 50(11): 1553-1564, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30349114

RESUMEN

The human genome encodes a variety of poorly understood RNA species that remain challenging to identify using existing genomic tools. We developed chromatin run-on and sequencing (ChRO-seq) to map the location of RNA polymerase for almost any input sample, including samples with degraded RNA that are intractable to RNA sequencing. We used ChRO-seq to map nascent transcription in primary human glioblastoma (GBM) brain tumors. Enhancers identified in primary GBMs resemble open chromatin in the normal human brain. Rare enhancers that are activated in malignant tissue drive regulatory programs similar to the developing nervous system. We identified enhancers that regulate groups of genes that are characteristic of each known GBM subtype and transcription factors that drive them. Finally we discovered a core group of transcription factors that control the expression of genes associated with clinical outcomes. This study characterizes the transcriptional landscape of GBM and introduces ChRO-seq as a method to map regulatory programs that contribute to complex diseases.


Asunto(s)
Neoplasias Encefálicas/genética , Mapeo Cromosómico/métodos , Glioblastoma/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Análisis de Secuencia de ARN/métodos , Transcriptoma/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/patología , Cromatina/genética , Cromatina/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Genoma Humano , Glioblastoma/patología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Células Jurkat , Desequilibrio de Ligamiento , Ratones , Ratones Desnudos , Elongación de la Transcripción Genética
8.
Nucleic Acids Res ; 46(21): 11502-11513, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30212902

RESUMEN

Gene expression programs change during cellular transitions. It is well established that a network of transcription factors and chromatin modifiers regulate RNA levels during embryonic stem cell (ESC) differentiation, but the full impact of post-transcriptional processes remains elusive. While cytoplasmic RNA turnover mechanisms have been implicated in differentiation, the contribution of nuclear RNA decay has not been investigated. Here, we differentiate mouse ESCs, depleted for the ribonucleolytic RNA exosome, into embryoid bodies to determine to which degree RNA abundance in the two states can be attributed to changes in transcription versus RNA decay by the exosome. As a general observation, we find that exosome depletion mainly leads to the stabilization of RNAs from lowly transcribed loci, including several protein-coding genes. Depletion of the nuclear exosome cofactor RBM7 leads to similar effects. In particular, transcripts that are differentially expressed between states tend to be more exosome sensitive in the state where expression is low. We conclude that the RNA exosome contributes to down-regulation of transcripts with disparate expression, often in conjunction with transcriptional down-regulation.


Asunto(s)
Diferenciación Celular/genética , Exosomas/genética , Regulación de la Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , ARN/genética , Animales , Exosomas/metabolismo , Perfilación de la Expresión Génica , Ratones , Células Madre Embrionarias de Ratones/citología , ARN/metabolismo , Interferencia de ARN , Estabilidad del ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Genes Dev ; 30(15): 1731-46, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27492368

RESUMEN

The coordinated regulation of gene expression at the transcriptional level is fundamental to development and homeostasis. Inducible systems are invaluable when studying transcription because the regulatory process can be triggered instantaneously, allowing the tracking of ordered mechanistic events. Here, we use precision run-on sequencing (PRO-seq) to examine the genome-wide heat shock (HS) response in Drosophila and the function of two key transcription factors on the immediate transcription activation or repression of all genes regulated by HS. We identify the primary HS response genes and the rate-limiting steps in the transcription cycle that GAGA-associated factor (GAF) and HS factor (HSF) regulate. We demonstrate that GAF acts upstream of promoter-proximally paused RNA polymerase II (Pol II) formation (likely at the step of chromatin opening) and that GAF-facilitated Pol II pausing is critical for HS activation. In contrast, HSF is dispensable for establishing or maintaining Pol II pausing but is critical for the release of paused Pol II into the gene body at a subset of highly activated genes. Additionally, HSF has no detectable role in the rapid HS repression of thousands of genes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , Regulación de la Expresión Génica/genética , Estrés Fisiológico/genética , Factores de Transcripción/metabolismo , Animales , Línea Celular , ADN Polimerasa II/metabolismo , Proteínas de Unión al ADN/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Factores de Transcripción del Choque Térmico , Regiones Promotoras Genéticas/genética , Interferencia de ARN , Factores de Transcripción/genética
10.
Nat Protoc ; 11(8): 1455-76, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27442863

RESUMEN

We provide a protocol for precision nuclear run-on sequencing (PRO-seq) and its variant, PRO-cap, which map the location of active RNA polymerases (PRO-seq) or transcription start sites (TSSs) (PRO-cap) genome-wide at high resolution. The density of RNA polymerases at a particular genomic locus directly reflects the level of nascent transcription at that region. Nuclei are isolated from cells and, under nuclear run-on conditions, transcriptionally engaged RNA polymerases incorporate one or, at most, a few biotin-labeled nucleotide triphosphates (biotin-NTPs) into the 3' end of nascent RNA. The biotin-labeled nascent RNA is used to prepare sequencing libraries, which are sequenced from the 3' end to provide high-resolution positional information for the RNA polymerases. PRO-seq provides much higher sensitivity than ChIP-seq, and it generates a much larger fraction of usable sequence reads than ChIP-seq or NET-seq (native elongating transcript sequencing). Similarly to NET-seq, PRO-seq maps the RNA polymerase at up to base-pair resolution with strand specificity, but unlike NET-seq it does not require immunoprecipitation. With the protocol provided here, PRO-seq (or PRO-cap) libraries for high-throughput sequencing can be generated in 4-5 working days. The method has been applied to human, mouse, Drosophila melanogaster and Caenorhabditis elegans cells and, with slight modifications, to yeast.


Asunto(s)
Emparejamiento Base , Mapeo Cromosómico/métodos , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN/química , ARN/genética , Animales , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Humanos , Ratones , ARN/metabolismo , Análisis de Secuencia de ARN , Sitio de Iniciación de la Transcripción
12.
Nat Methods ; 12(5): 433-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25799441

RESUMEN

Modifications to the global run-on and sequencing (GRO-seq) protocol that enrich for 5'-capped RNAs can be used to reveal active transcriptional regulatory elements (TREs) with high accuracy. Here, we introduce discriminative regulatory-element detection from GRO-seq (dREG), a sensitive machine learning method that uses support vector regression to identify active TREs from GRO-seq data without requiring cap-based enrichment (https://github.com/Danko-Lab/dREG/). This approach allows TREs to be assayed together with gene expression levels and other transcriptional features in a single experiment. Predicted TREs are more enriched for several marks of transcriptional activation­including expression quantitative trait loci, disease-associated polymorphisms, acetylated histone 3 lysine 27 (H3K27ac) and transcription factor binding­than those identified by alternative functional assays. Using dREG, we surveyed TREs in eight human cell types and provide new insights into global patterns of TRE function.


Asunto(s)
Inteligencia Artificial , Regulación de la Expresión Génica/fisiología , Elementos Reguladores de la Transcripción/fisiología , Línea Celular , Estudio de Asociación del Genoma Completo , Histonas , Humanos , Células K562 , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Elementos Reguladores de la Transcripción/genética , Programas Informáticos
13.
Nat Commun ; 5: 5336, 2014 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-25387874

RESUMEN

Mammalian genomes are pervasively transcribed, yielding a complex transcriptome with high variability in composition and cellular abundance. Although recent efforts have identified thousands of new long non-coding (lnc) RNAs and demonstrated a complex transcriptional repertoire produced by protein-coding (pc) genes, limited progress has been made in distinguishing functional RNA from spurious transcription events. This is partly due to present RNA classification, which is typically based on technical rather than biochemical criteria. Here we devise a strategy to systematically categorize human RNAs by their sensitivity to the ribonucleolytic RNA exosome complex and by the nature of their transcription initiation. These measures are surprisingly effective at correctly classifying annotated transcripts, including lncRNAs of known function. The approach also identifies uncharacterized stable lncRNAs, hidden among a vast majority of unstable transcripts. The predictive power of the approach promises to streamline the functional analysis of known and novel RNAs.


Asunto(s)
Estabilidad del ARN/fisiología , ARN/clasificación , Exosomas/genética , Exosomas/fisiología , Células HeLa , Humanos , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , ARN/fisiología , Estabilidad del ARN/genética , ARN no Traducido/fisiología , Iniciación de la Transcripción Genética/fisiología , Transcripción Genética/fisiología
14.
Nat Genet ; 46(12): 1311-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25383968

RESUMEN

Despite the conventional distinction between them, promoters and enhancers share many features in mammals, including divergent transcription and similar modes of transcription factor binding. Here we examine the architecture of transcription initiation through comprehensive mapping of transcription start sites (TSSs) in human lymphoblastoid B cell (GM12878) and chronic myelogenous leukemic (K562) ENCODE Tier 1 cell lines. Using a nuclear run-on protocol called GRO-cap, which captures TSSs for both stable and unstable transcripts, we conduct detailed comparisons of thousands of promoters and enhancers in human cells. These analyses identify a common architecture of initiation, including tightly spaced (110 bp apart) divergent initiation, similar frequencies of core promoter sequence elements, highly positioned flanking nucleosomes and two modes of transcription factor binding. Post-initiation transcript stability provides a more fundamental distinction between promoters and enhancers than patterns of histone modification and association of transcription factors or co-activators. These results support a unified model of transcription initiation at promoters and enhancers.


Asunto(s)
Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , ARN/genética , Linfocitos B/citología , Sitios de Unión , Cromatina/química , Histonas/química , Humanos , Células K562 , Cadenas de Markov , Modelos Genéticos , Nucleosomas/química , Empalme del ARN , Secuencias Reguladoras de Ácidos Nucleicos , Sitio de Iniciación de la Transcripción , Transcripción Genética
15.
Cell Rep ; 6(5): 906-15, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24561252

RESUMEN

RNA sequences are expected to be identical to their corresponding DNA sequences. Here, we found all 12 types of RNA-DNA sequence differences (RDDs) in nascent RNA. Our results show that RDDs begin to occur in RNA chains ~55 nt from the RNA polymerase II (Pol II) active site. These RDDs occur so soon after transcription that they are incompatible with known deaminase-mediated RNA-editing mechanisms. Moreover, the 55 nt delay in appearance indicates that they do not arise during RNA synthesis by Pol II or as a direct consequence of modified base incorporation. Preliminary data suggest that RDD and R-loop formations may be coupled. These findings identify sequence substitution as an early step in cotranscriptional RNA processing.


Asunto(s)
ADN/metabolismo , ARN Polimerasa II/metabolismo , ARN/metabolismo , Dominio Catalítico , Técnicas de Cultivo de Célula , ADN/genética , Expresión Génica , Humanos , ARN/genética , Transcripción Genética
16.
Cell Rep ; 6(3): 455-66, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24485661

RESUMEN

Fluctuations in nutrient availability profoundly impact gene expression. Previous work revealed postrecruitment regulation of RNA polymerase II (Pol II) during starvation and recovery in Caenorhabditis elegans, suggesting that promoter-proximal pausing promotes rapid response to feeding. To test this hypothesis, we measured Pol II elongation genome wide by two complementary approaches and analyzed elongation in conjunction with Pol II binding and expression. We confirmed bona fide pausing during starvation and also discovered Pol II docking. Pausing occurs at active stress-response genes that become downregulated in response to feeding. In contrast, "docked" Pol II accumulates without initiating upstream of inactive growth genes that become rapidly upregulated upon feeding. Beyond differences in function and expression, these two sets of genes have different core promoter motifs, suggesting alternative transcriptional machinery. Our work suggests that growth and stress genes are both regulated postrecruitment during starvation but at initiation and elongation, respectively, coordinating gene expression with nutrient availability.


Asunto(s)
Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/genética , Genes de Helminto/genética , ARN Polimerasa II/metabolismo , Estrés Fisiológico/genética , Animales , Inmunoprecipitación de Cromatina , Regulación del Desarrollo de la Expresión Génica , Mutación/genética , Regiones Promotoras Genéticas , Caperuzas de ARN/genética , ARN de Helminto/genética , ARN de Helminto/metabolismo , Análisis de Secuencia de ARN , Sitio de Iniciación de la Transcripción , Transcripción Genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
17.
Science ; 342(6159): 744-7, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24136355

RESUMEN

DNA sequence variation has been associated with quantitative changes in molecular phenotypes such as gene expression, but its impact on chromatin states is poorly characterized. To understand the interplay between chromatin and genetic control of gene regulation, we quantified allelic variability in transcription factor binding, histone modifications, and gene expression within humans. We found abundant allelic specificity in chromatin and extensive local, short-range, and long-range allelic coordination among the studied molecular phenotypes. We observed genetic influence on most of these phenotypes, with histone modifications exhibiting strong context-dependent behavior. Our results implicate transcription factors as primary mediators of sequence-specific regulation of gene expression programs, with histone modifications frequently reflecting the primary regulatory event.


Asunto(s)
Cromatina/metabolismo , ADN/metabolismo , Regulación de la Expresión Génica , Variación Genética , Factores de Transcripción/metabolismo , Transcripción Genética , Alelos , Secuencia de Bases/genética , Sitios de Unión/genética , Cromatina/química , ADN/química , Histonas/química , Histonas/metabolismo , Humanos , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas
18.
Elife ; 2: e00808, 2013 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-23795297

RESUMEN

The X-chromosome gene regulatory process called dosage compensation ensures that males (1X) and females (2X) express equal levels of X-chromosome transcripts. The mechanism in Caenorhabditis elegans has been elusive due to improperly annotated transcription start sites (TSSs). Here we define TSSs and the distribution of transcriptionally engaged RNA polymerase II (Pol II) genome-wide in wild-type and dosage-compensation-defective animals to dissect this regulatory mechanism. Our TSS-mapping strategy integrates GRO-seq, which tracks nascent transcription, with a new derivative of this method, called GRO-cap, which recovers nascent RNAs with 5' caps prior to their removal by co-transcriptional processing. Our analyses reveal that promoter-proximal pausing is rare, unlike in other metazoans, and promoters are unexpectedly far upstream from the 5' ends of mature mRNAs. We find that C. elegans equalizes X-chromosome expression between the sexes, to a level equivalent to autosomes, by reducing Pol II recruitment to promoters of hermaphrodite X-linked genes using a chromosome-restructuring condensin complex. DOI:http://dx.doi.org/10.7554/eLife.00808.001.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Proteínas de Unión al ADN/fisiología , Compensación de Dosificación (Genética) , Complejos Multiproteicos/fisiología , Nematodos/genética , ARN Polimerasa II/metabolismo , Cromosoma X , Animales , Genoma , Regiones Promotoras Genéticas , Transcripción Genética
19.
Genes Dev ; 27(10): 1146-58, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23699410

RESUMEN

Cascades of zygotic gene expression pattern the anterior-posterior (AP) and dorsal-ventral (DV) axes of the early Drosophila embryo. Here, we used the global run-on sequencing assay (GRO-seq) to map the genome-wide RNA polymerase distribution during early Drosophila embryogenesis, thus providing insights into how genes are regulated. We identify widespread promoter-proximal pausing yet show that the presence of paused polymerase does not necessarily equate to direct regulation through pause release to productive elongation. Our data reveal that a subset of early Zelda-activated genes is regulated at the level of polymerase recruitment, whereas other Zelda target and axis patterning genes are predominantly regulated through pause release. In contrast to other signaling pathways, we found that bone morphogenetic protein (BMP) target genes are collectively more highly paused than BMP pathway components and show that BMP target gene expression requires the pause-inducing negative elongation factor (NELF) complex. Our data also suggest that polymerase pausing allows plasticity in gene activation throughout embryogenesis, as transiently repressed and transcriptionally silenced genes maintain and lose promoter polymerases, respectively. Finally, we provide evidence that the major effect of pausing is on the levels, rather than timing, of transcription. These data are discussed in terms of the efficiency of transcriptional activation required across cell populations during developmental time constraints.


Asunto(s)
Tipificación del Cuerpo/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , ARN Polimerasa II/metabolismo , Transcripción Genética , Animales , Proteínas de Drosophila/metabolismo , Femenino , Proteínas Nucleares , Factores de Transcripción/metabolismo , Cigoto/metabolismo
20.
Mol Cell ; 50(2): 212-22, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23523369

RESUMEN

RNA polymerase II (Pol II) transcribes hundreds of kilobases of DNA, limiting the production of mRNAs and lncRNAs. We used global run-on sequencing (GRO-seq) to measure the rates of transcription by Pol II following gene activation. Elongation rates vary as much as 4-fold at different genomic loci and in response to two distinct cellular signaling pathways (i.e., 17ß-estradiol [E2] and TNF-α). The rates are slowest near the promoter and increase during the first ~15 kb transcribed. Gene body elongation rates correlate with Pol II density, resulting in systematically higher rates of transcript production at genes with higher Pol II density. Pol II dynamics following short inductions indicate that E2 stimulates gene expression by increasing Pol II initiation, whereas TNF-α reduces Pol II residence time at pause sites. Collectively, our results identify previously uncharacterized variation in the rate of transcription and highlight elongation as an important, variable, and regulated rate-limiting step during transcription.


Asunto(s)
ARN Polimerasa II/metabolismo , ARN Mensajero/biosíntesis , Transducción de Señal , Iniciación de la Transcripción Genética , Estradiol/farmacología , Estradiol/fisiología , Humanos , Cinética , Células MCF-7 , Regiones Promotoras Genéticas , ARN Polimerasa II/fisiología , ARN Mensajero/genética , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética , Activación Transcripcional , Transcriptoma , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...