Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bio Protoc ; 13(18): e4828, 2023 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-37753464

RESUMEN

In eukaryotic cells, RNA biogenesis generally requires processing of the nascent transcript as it is being synthesized by RNA polymerase. These processing events include endonucleolytic cleavage, exonucleolytic trimming, and splicing of the growing nascent transcript. Endonucleolytic cleavage events that generate an exposed 5'-monophosphorylated (5'-PO4) end on the growing nascent transcript occur in the maturation of rRNAs, tRNAs, and mRNAs. These 5'-PO4 ends can be a target of further processing or be subjected to 5'-3' exonucleolytic digestion that may result in termination of transcription. Here, we describe how to identify 5'-PO4 ends of intermediates in nascent RNA metabolism. We capture these species via metabolic labeling with bromouridine followed by immunoprecipitation and specific ligation of 5'-PO4 RNA ends with the 3'-hydroxyl group of a 5' adaptor (5'-PO4 Bru-Seq) using RNA ligase I. These ligation events are localized at single nucleotide resolution via highthroughput sequencing, which identifies the position of 5'-PO4 groups precisely. This protocol successfully detects the 5'monophosphorylated ends of RNA processing intermediates during production of mature ribosomal, transfer, and micro RNAs. When combined with inhibition of the nuclear 5'-3' exonuclease Xrn2, 5'-PO4 Bru-Seq maps the 5' splice sites of debranched introns and mRNA and tRNA 3' end processing sites cleaved by CPSF73 and RNaseZ, respectively. Key features • Metabolic labeling for brief periods with bromouridine focuses the analysis of 5'-PO4 RNA ends on the population of nascent transcripts that are actively transcribed. • Detects 5'-PO4 RNA ends on nascent transcripts produced by all RNA polymerases. • Detects 5'-PO4 RNA ends at single nucleotide resolution.

2.
Elife ; 112022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36421680

RESUMEN

Alternative polyadenylation yields many mRNA isoforms whose 3' termini occur disproportionately in clusters within 3' untranslated regions. Previously, we showed that profiles of poly(A) site usage are regulated by the rate of transcriptional elongation by RNA polymerase (Pol) II (Geisberg et al., 2020). Pol II derivatives with slow elongation rates confer an upstream-shifted poly(A) profile, whereas fast Pol II strains confer a downstream-shifted poly(A) profile. Within yeast isoform clusters, these shifts occur steadily from one isoform to the next across nucleotide distances. In contrast, the shift between clusters - from the last isoform of one cluster to the first isoform of the next - is much less pronounced, even over large distances. GC content in a region 13-30 nt downstream from isoform clusters correlates with their sensitivity to Pol II elongation rate. In human cells, the upstream shift caused by a slow Pol II mutant also occurs continuously at single nucleotide resolution within clusters but not between them. Pol II occupancy increases just downstream of poly(A) sites, suggesting a linkage between reduced elongation rate and cluster formation. These observations suggest that (1) Pol II elongation speed affects the nucleotide-level dwell time allowing polyadenylation to occur, (2) poly(A) site clusters are linked to the local elongation rate, and hence do not arise simply by intrinsically imprecise cleavage and polyadenylation of the RNA substrate, (3) DNA sequence elements can affect Pol II elongation and poly(A) profiles, and (4) the cleavage/polyadenylation and Pol II elongation complexes are spatially, and perhaps physically, coupled so that polyadenylation occurs rapidly upon emergence of the nascent RNA from the Pol II elongation complex.


Asunto(s)
Nucleótidos , Poliadenilación , Humanos , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Poli A/genética , Poli A/metabolismo , Saccharomyces cerevisiae/genética , Regiones no Traducidas 3' , Transcripción Genética
3.
Genes Dev ; 36(19-20): 1062-1078, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36396340

RESUMEN

The exonuclease torpedo Xrn2 loads onto nascent RNA 5'-PO4 ends and chases down pol II to promote termination downstream from polyA sites. We report that Xrn2 is recruited to preinitiation complexes and "travels" to 3' ends of genes. Mapping of 5'-PO4 ends in nascent RNA identified Xrn2 loading sites stabilized by an active site mutant, Xrn2(D235A). Xrn2 loading sites are approximately two to 20 bases downstream from where CPSF73 cleaves at polyA sites and histone 3' ends. We propose that processing of all mRNA 3' ends comprises cleavage and limited 5'-3' trimming by CPSF73, followed by handoff to Xrn2. A similar handoff occurs at tRNA 3' ends, where cotranscriptional RNase Z cleavage generates novel Xrn2 substrates. Exonuclease-dead Xrn2 increased transcription in 3' flanking regions by inhibiting polyA site-dependent termination. Surprisingly, the mutant Xrn2 also rescued transcription in promoter-proximal regions to the same extent as in 3' flanking regions. eNET-seq revealed Xrn2-mediated degradation of sense and antisense nascent RNA within a few bases of the TSS, where 5'-PO4 ends may be generated by decapping or endonucleolytic cleavage. These results suggest that a major fraction of pol II complexes terminates prematurely close to the start site under normal conditions by an Xrn2-mediated torpedo mechanism.


Asunto(s)
Poli A , ARN Polimerasa II , ARN Polimerasa II/genética , Núcleo Celular , Exonucleasas , ARN sin Sentido
4.
Mol Cell ; 82(19): 3632-3645.e4, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36206739

RESUMEN

The pause-release model of transcription proposes that 40-100 bases from the start site RNA Pol II pauses, followed by release into productive elongation. Pause release is facilitated by the PTEFb phosphorylation of the RNA Pol II elongation factor, Spt5. We mapped paused polymerases by eNET-seq and found frequent pausing in zones that extend ∼0.3-3 kb into genes even when PTEFb is inhibited. The fraction of paused polymerases or pausing propensity declines gradually over several kb and not abruptly as predicted for a discrete pause-release event. Spt5 depletion extends pausing zones, suggesting that it promotes the maturation of elongation complexes to a low-pausing state. The expression of mutants after Spt5 depletion showed that phosphomimetic substitutions in the CTR1 domain diminished pausing throughout genes. By contrast, mutants that prevent the phosphorylation of the Spt5 RNA-binding domain strengthened pausing. Thus, distinct Spt5 phospho-isoforms set the balance between pausing and elongation.


Asunto(s)
ARN Polimerasa II , Factores de Elongación Transcripcional , Factores de Elongación de Péptidos/metabolismo , Fosforilación , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Transcripción Genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
5.
BMC Genomics ; 22(1): 476, 2021 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-34174817

RESUMEN

BACKGROUND: The sequence content of the 3' UTRs of many mRNA transcripts is regulated through alternative polyadenylation (APA). The study of this process using RNAseq data, though, has been historically challenging. RESULTS: To combat this problem, we developed LABRAT, an APA isoform quantification method. LABRAT takes advantage of newly developed transcriptome quantification techniques to accurately determine relative APA site usage and how it varies across conditions. Using LABRAT, we found consistent relationships between gene-distal APA and subcellular RNA localization in multiple cell types. We also observed connections between transcription speed and APA site choice as well as tumor-specific transcriptome-wide shifts in APA isoform abundance in hundreds of patient-derived tumor samples that were associated with patient prognosis. We investigated the effects of APA on transcript expression and found a weak overall relationship, although many individual genes showed strong correlations between relative APA isoform abundance and overall gene expression. We interrogated the roles of 191 RNA-binding proteins in the regulation of APA isoforms, finding that dozens promote broad, directional shifts in relative APA isoform abundance both in vitro and in patient-derived samples. Finally, we find that APA site shifts in the two classes of APA, tandem UTRs and alternative last exons, are strongly correlated across many contexts, suggesting that they are coregulated. CONCLUSIONS: We conclude that LABRAT has the ability to accurately quantify APA isoform ratios from RNAseq data across a variety of sample types. Further, LABRAT is able to derive biologically meaningful insights that connect APA isoform regulation to cellular and molecular phenotypes.


Asunto(s)
Neoplasias , Poliadenilación , Regiones no Traducidas 3' , Humanos , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética
6.
Proc Natl Acad Sci U S A ; 117(33): 19888-19895, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32747552

RESUMEN

More than 30% of genes in higher eukaryotes are regulated by RNA polymerase II (Pol II) promoter proximal pausing. Pausing is released by the positive transcription elongation factor complex (P-TEFb). However, the exact mechanism by which this occurs and whether phosphorylation of the carboxyl-terminal domain of Pol II is involved in the process remains unknown. We previously reported that JMJD5 could generate tailless nucleosomes at position +1 from transcription start sites (TSS), thus perhaps enable progression of Pol II. Here we find that knockout of JMJD5 leads to accumulation of nucleosomes at position +1. Absence of JMJD5 also results in loss of or lowered transcription of a large number of genes. Interestingly, we found that phosphorylation, by CDK9, of Ser2 within two neighboring heptad repeats in the carboxyl-terminal domain of Pol II, together with phosphorylation of Ser5 within the second repeat, HR-Ser2p (1, 2)-Ser5p (2) for short, allows Pol II to bind JMJD5 via engagement of the N-terminal domain of JMJD5. We suggest that these events bring JMJD5 near the nucleosome at position +1, thus allowing JMJD5 to clip histones on this nucleosome, a phenomenon that may contribute to release of Pol II pausing.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Histona Demetilasas/metabolismo , ARN Polimerasa II/metabolismo , Transcripción Genética , Línea Celular Tumoral , Quinasa 9 Dependiente de la Ciclina/genética , Histona Demetilasas/química , Histona Demetilasas/genética , Humanos , Nucleosomas/genética , Nucleosomas/metabolismo , Fosforilación , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Dominios Proteicos , ARN Polimerasa II/genética
7.
Mol Cell ; 76(6): 896-908.e4, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31677974

RESUMEN

Control of transcription speed, which influences many co-transcriptional processes, is poorly understood. We report that PNUTS-PP1 phosphatase is a negative regulator of RNA polymerase II (Pol II) elongation rate. The PNUTS W401A mutation, which disrupts PP1 binding, causes genome-wide acceleration of transcription associated with hyper-phosphorylation of the Spt5 elongation factor. Immediately downstream of poly(A) sites, Pol II decelerates from >2 kb/min to <1 kb/min, which correlates with Spt5 dephosphorylation. Pol II deceleration and Spt5 dephosphorylation require poly(A) site recognition and the PNUTS-PP1 complex, which is in turn necessary for transcription termination. These results lead to a model for termination, the "sitting duck torpedo" mechanism, where poly(A) site-dependent deceleration caused by PNUTS-PP1 and Spt5 dephosphorylation is required to convert Pol II into a viable target for the Xrn2 terminator exonuclease. Spt5 and its bacterial homolog NusG therefore have related functions controlling kinetic competition between RNA polymerases and the termination factors that pursue them.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Exorribonucleasas/metabolismo , Proteína Fosfatasa 1/metabolismo , Procesamiento Proteico-Postraduccional , ARN Polimerasa II/metabolismo , ARN Mensajero/biosíntesis , Proteínas de Unión al ARN/metabolismo , Terminación de la Transcripción Genética , Sitios de Unión , Proteínas de Unión al ADN/genética , Exorribonucleasas/genética , Células HEK293 , Humanos , Cinética , Proteínas Nucleares/genética , Fosforilación , Poli A/metabolismo , Unión Proteica , Proteína Fosfatasa 1/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Transducción de Señal , Factores de Elongación Transcripcional/genética
8.
Mol Cell ; 73(1): 107-118.e4, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30503775

RESUMEN

In addition to phosphodiester bond formation, RNA polymerase II has an RNA endonuclease activity, stimulated by TFIIS, which rescues complexes that have arrested and backtracked. How TFIIS affects transcription under normal conditions is poorly understood. We identified backtracking sites in human cells using a dominant-negative TFIIS (TFIISDN) that inhibits RNA cleavage and stabilizes backtracked complexes. Backtracking is most frequent within 2 kb of start sites, consistent with slow elongation early in transcription, and in 3' flanking regions where termination is enhanced by TFIISDN, suggesting that backtracked pol II is a favorable substrate for termination. Rescue from backtracking by RNA cleavage also promotes escape from 5' pause sites, prevents premature termination of long transcripts, and enhances activation of stress-inducible genes. TFIISDN slowed elongation rates genome-wide by half, suggesting that rescue of backtracked pol II by TFIIS is a major stimulus of elongation under normal conditions.


Asunto(s)
División del ARN , ARN Polimerasa II/metabolismo , ARN/metabolismo , Elongación de la Transcripción Genética , Terminación de la Transcripción Genética , Activación Transcripcional , Región de Flanqueo 3' , Animales , Regulación de la Expresión Génica , Células HEK293 , Humanos , Cinética , Ratones , Mutación , ARN/genética , ARN Polimerasa II/genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
10.
Genes Dev ; 32(3-4): 297-308, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29483154

RESUMEN

Transcription elongation rate influences cotranscriptional pre-mRNA maturation, but how such kinetic coupling works is poorly understood. The formation of nonadenylated histone mRNA 3' ends requires recognition of an RNA structure by stem-loop-binding protein (SLBP). We report that slow transcription by mutant RNA polymerase II (Pol II) caused accumulation of polyadenylated histone mRNAs that extend past the stem-loop processing site. UV irradiation, which decelerates Pol II elongation, also induced long poly(A)+ histone transcripts. Inhibition of 3' processing by slow Pol II correlates with failure to recruit SLBP to histone genes. Chemical probing of nascent RNA structure showed that the stem-loop fails to fold in transcripts made by slow Pol II, thereby explaining the absence of SLBP and failure to process 3' ends. These results show that regulation of transcription speed can modulate pre-mRNA processing by changing nascent RNA structure and suggest a mechanism by which alternative processing could be controlled.


Asunto(s)
Histonas/genética , Procesamiento de Término de ARN 3' , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Elongación de la Transcripción Genética , Células HEK293 , Histonas/metabolismo , Humanos , Cinética , Proteínas Nucleares/metabolismo , Pliegue del ARN , Precursores del ARN/química , ARN Mensajero/química , Transcripción Genética/efectos de la radiación , Rayos Ultravioleta , Factores de Escisión y Poliadenilación de ARNm/metabolismo
11.
Cell Rep ; 20(5): 1173-1186, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28768201

RESUMEN

CDK7 phosphorylates the RNA polymerase II (pol II) C-terminal domain CTD and activates the P-TEFb-associated kinase CDK9, but its regulatory roles remain obscure. Here, using human CDK7 analog-sensitive (CDK7as) cells, we observed reduced capping enzyme recruitment, increased pol II promoter-proximal pausing, and defective termination at gene 3' ends upon CDK7 inhibition. We also noted that CDK7 regulates chromatin modifications downstream of transcription start sites. H3K4me3 spreading was restricted at gene 5' ends and H3K36me3 was displaced toward gene 3' ends in CDK7as cells. Mass spectrometry identified factors that bound TFIIH-phosphorylated versus P-TEFb-phosphorylated CTD (versus unmodified); capping enzymes and H3K4 methyltransferase complexes, SETD1A/B, selectively bound phosphorylated CTD, and the H3K36 methyltransferase SETD2 specifically bound P-TEFb-phosphorylated CTD. Moreover, TFIIH-phosphorylated CTD stimulated SETD1A/B activity toward nucleosomes, revealing a mechanistic basis for CDK7 regulation of H3K4me3 spreading. Collectively, these results implicate a CDK7-dependent "CTD code" that regulates chromatin marks in addition to RNA processing and pol II pausing.


Asunto(s)
Cromatina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Factor de Transcripción TFIIH/metabolismo , Transcripción Genética , Línea Celular , Cromatina/genética , Quinasas Ciclina-Dependientes/genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Fosforilación , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Factor de Transcripción TFIIH/genética , Quinasa Activadora de Quinasas Ciclina-Dependientes
12.
Mol Cell ; 66(4): 546-557.e3, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28506463

RESUMEN

Eukaryotic genes are marked by conserved post-translational modifications on the RNA pol II C-terminal domain (CTD) and the chromatin template. How the 5'-3' profiles of these marks are established is poorly understood. Using pol II mutants in human cells, we found that slow transcription repositioned specific co-transcriptionally deposited chromatin modifications; histone H3 lysine 36 trimethyl (H3K36me3) shifted within genes toward 5' ends, and histone H3 lysine 4 dimethyl (H3K4me2) extended farther upstream of start sites. Slow transcription also evoked a hyperphosphorylation of CTD Ser2 residues at 5' ends of genes that is conserved in yeast. We propose a "dwell time in the target zone" model to explain the effects of transcriptional dynamics on the establishment of co-transcriptionally deposited protein modifications. Promoter-proximal Ser2 phosphorylation is associated with a longer pol II dwell time at start sites and reduced transcriptional polarity because of strongly enhanced divergent antisense transcription at promoters. These results demonstrate that pol II dynamics help govern the decision between sense and divergent antisense transcription.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/enzimología , ADN de Hongos/metabolismo , ARN Polimerasa II/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Transcripción Genética , Cromatina/genética , ADN de Hongos/genética , Regulación Fúngica de la Expresión Génica , Células HEK293 , Humanos , Mutación , Fosforilación , Dominios Proteicos , ARN Polimerasa II/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Factores de Tiempo
13.
Mol Cell ; 60(2): 256-67, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26474067

RESUMEN

The torpedo model of transcription termination asserts that the exonuclease Xrn2 attacks the 5'PO4-end exposed by nascent RNA cleavage and chases down the RNA polymerase. We tested this mechanism using a dominant-negative human Xrn2 mutant and found that it delayed termination genome-wide. Xrn2 nuclease inactivation caused strong termination defects downstream of most poly(A) sites and modest delays at some histone and U snRNA genes, suggesting that the torpedo mechanism is not limited to poly(A) site-dependent termination. A central untested feature of the torpedo model is that there is kinetic competition between the exonuclease and the pol II elongation complex. Using pol II rate mutants, we found that slow transcription robustly shifts termination upstream, and fast elongation extends the zone of termination further downstream. These results suggest that kinetic competition between elongating pol II and the Xrn2 exonuclease is integral to termination of transcription on most human genes.


Asunto(s)
Exorribonucleasas/genética , Poli A/genética , ARN Polimerasa II/genética , ARN Mensajero/genética , Elongación de la Transcripción Genética , Terminación de la Transcripción Genética , Línea Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Exorribonucleasas/metabolismo , Genoma Humano , Células HEK293 , Células HeLa , Humanos , Cinética , Linfocitos/citología , Linfocitos/metabolismo , Modelos Genéticos , Mutación , Poli A/metabolismo , ARN Polimerasa II/metabolismo , ARN Mensajero/metabolismo
14.
Genes Dev ; 28(23): 2663-76, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25452276

RESUMEN

Alternative splicing modulates expression of most human genes. The kinetic model of cotranscriptional splicing suggests that slow elongation expands and that fast elongation compresses the "window of opportunity" for recognition of upstream splice sites, thereby increasing or decreasing inclusion of alternative exons. We tested the model using RNA polymerase II mutants that change average elongation rates genome-wide. Slow and fast elongation affected constitutive and alternative splicing, frequently altering exon inclusion and intron retention in ways not predicted by the model. Cassette exons included by slow and excluded by fast elongation (type I) have weaker splice sites, shorter flanking introns, and distinct sequence motifs relative to "slow-excluded" and "fast-included" exons (type II). Many rate-sensitive exons are misspliced in tumors. Unexpectedly, slow and fast elongation often both increased or both decreased inclusion of a particular exon or retained intron. These results suggest that an optimal rate of transcriptional elongation is required for normal cotranscriptional pre-mRNA splicing.


Asunto(s)
ARN Polimerasa II/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN , Elongación de la Transcripción Genética/fisiología , Exones/genética , Células HEK293 , Humanos , Intrones/genética , Mutación , ARN Polimerasa II/genética , Precursores del ARN/genética
15.
Mol Cell ; 46(3): 311-24, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22483619

RESUMEN

We report a function of human mRNA decapping factors in control of transcription by RNA polymerase II. Decapping proteins Edc3, Dcp1a, and Dcp2 and the termination factor TTF2 coimmunoprecipitate with Xrn2, the nuclear 5'-3' exonuclease "torpedo" that facilitates transcription termination at the 3' ends of genes. Dcp1a, Xrn2, and TTF2 localize near transcription start sites (TSSs) by ChIP-seq. At genes with 5' peaks of paused pol II, knockdown of decapping or termination factors Xrn2 and TTF2 shifted polymerase away from the TSS toward upstream and downstream distal positions. This redistribution of pol II is similar in magnitude to that caused by depletion of the elongation factor Spt5. We propose that coupled decapping of nascent transcripts and premature termination by the "torpedo" mechanism is a widespread mechanism that limits bidirectional pol II elongation. Regulated cotranscriptional decapping near promoter-proximal pause sites followed by premature termination could control productive pol II elongation.


Asunto(s)
Exorribonucleasas/fisiología , ARN Polimerasa II/fisiología , Estabilidad del ARN , ARN Mensajero/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Células HEK293 , Células HeLa , Humanos , Modelos Genéticos , Mapeo de Interacción de Proteínas , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Transcripción Genética
16.
Proc Natl Acad Sci U S A ; 108(33): 13564-9, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21807997

RESUMEN

A chromatin code appears to mark introns and exons with distinct patterns of nucleosome enrichment and histone methylation. We investigated whether a causal relationship exists between splicing and chromatin modification by asking whether splice-site mutations affect the methylation of histone H3K36. Deletions of the 3' splice site in intron 2 or in both introns 1 and 2 of an integrated ß-globin reporter gene caused a shift in relative distribution of H3K36 trimethylation away from 5' ends and toward 3' ends. The effects of splice-site mutations correlated with enhanced retention of a U5 snRNP subunit on transcription complexes downstream of the gene. In contrast, a poly(A) site mutation did not affect H3K36 methylation. Similarly, global inhibition of splicing by spliceostatin A caused a rapid repositioning of H3K36me3 away from 5' ends in favor of 3' ends. These results suggest that the cotranscriptional splicing apparatus influences establishment of normal patterns of histone modification.


Asunto(s)
Histonas/metabolismo , Precursores del ARN/metabolismo , Empalme del ARN , Línea Celular , Histonas/genética , Humanos , Metilación , Mutación , Procesamiento Proteico-Postraduccional , Sitios de Empalme de ARN/genética , ARN Mensajero , Ribonucleoproteínas Nucleares Pequeñas , Globinas beta/genética
17.
Nat Struct Mol Biol ; 16(9): 916-22, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19701200

RESUMEN

We investigated whether a continuous transcript is necessary for co-transcriptional pre-mRNA processing. Cutting an intron with the fast-cleaving hepatitis delta ribozyme, but not the slower hammerhead, inhibited splicing. Therefore, exon tethering to RNA polymerase II (Pol II) cannot rescue splicing of a transcript severed by a ribozyme that cleaves rapidly relative to the rate of splicing. Ribozyme cutting also released cap-binding complex (CBC) from the gene, suggesting that exon 1 is not tethered. Unexpectedly, cutting within exons inhibited splicing of distal introns, where exon definition is not affected, probably owing to disruption of the interactions with the CBC and the Pol II C-terminal domain that facilitate splicing. Ribozyme cutting within the mRNA also inhibited 3' processing and transcription termination. We propose that damaging the nascent transcript aborts pre-mRNA processing and that this mechanism may help to prevent association of processing factors with Pol II that is not productively engaged in transcription.


Asunto(s)
ARN Polimerasa II/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN Catalítico/metabolismo , Transcripción Genética , Animales , Células CHO , Cricetinae , Cricetulus , Exones , Virus de la Hepatitis Delta/enzimología , Intrones , Edición de ARN , Empalme del ARN
18.
RNA ; 13(7): 1071-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17525170

RESUMEN

The C-terminal domain (CTD) of the large subunit of RNA polymerase II (Pol II) influences many steps in the synthesis of an mRNA and helps control the final destiny of the mature transcript. ADAR2 edits RNA by converting adenosine to inosine within double-stranded or structured RNA. Site-selective A-to-I editing often occurs at sites near exon/intron borders, where it depends on intronic sequences for substrate recognition. It is therefore essential that editing precedes splicing. We have investigated whether there is coordination between ADAR2 editing and splicing of the GluR-B pre-mRNA. We show that the CTD is required for efficient editing at the R/G site one base upstream of a 5'-splice site. The results suggest that the CTD enhances editing at the R/G site by preventing premature splicing that would remove the intronic recognition sites for ADAR2. Editing at the GluR-B Q/R site, 24 bases upstream of the intron 11 5'-splice site, stimulates splicing at this intron. Furthermore, unlike previously studied introns, the CTD actually inhibits excision of intron 11, which includes the complementary recognition sequences for the Q/R editing site. In summary, these results show that the CTD and ADAR2 function together to enforce the order of events that allows editing to precede splicing, and they furthermore suggest a new role for the CTD as a coordinator of two interdependent pre-mRNA processing events.


Asunto(s)
Edición de ARN , ARN Polimerasa II/fisiología , Precursores del ARN/metabolismo , Empalme del ARN , Receptores AMPA/genética , Adenosina Desaminasa/metabolismo , Secuencia de Bases , Células Cultivadas , Eliminación de Gen , Humanos , Modelos Biológicos , Estructura Terciaria de Proteína/fisiología , ARN Polimerasa II/química , ARN Polimerasa II/genética , Sitios de Empalme de ARN , Proteínas de Unión al ARN , Receptores AMPA/metabolismo , Factores de Tiempo , Transfección
19.
EMBO Rep ; 7(3): 303-7, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16440002

RESUMEN

The carboxy-terminal domain (CTD) of the large subunit of RNA polymerase II (pol II) is essential for several co-transcriptional pre-messenger RNA processing events, including capping, 3'-end processing and splicing. We investigated the role of the CTD of RNA pol II in the coordination of A to I editing and splicing of the ADAR2 (ADAR: adenosine deaminases that act on RNA) pre-mRNA. The auto-editing of Adar2 intron 4 by the ADAR2 adenosine deaminase is tightly coupled to splicing, as the modification of the dinucleotide AA to AI creates a new 3' splice site. Unlike other introns, the CTD is not required for efficient splicing of intron 4 at either the normal 3' splice site or the alternative site created by editing. However, the CTD is required for efficient co-transcriptional auto-editing of ADAR2 intron 4. Our results implicate the CTD in site-selective RNA editing by ADAR2 and in coordination of editing with alternative splicing.


Asunto(s)
Adenosina Desaminasa , Empalme Alternativo , Edición de ARN , ARN/metabolismo , Transcripción Genética , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Secuencia de Bases , Humanos , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN/genética , ARN Polimerasa II/metabolismo , Proteínas de Unión al ARN , Ratas
20.
Mol Cell ; 20(5): 747-58, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16337598

RESUMEN

We report a functional connection between splicing and transcript release from the DNA. A Pol II CTD mutant inhibited not only splicing but also RNA release from the site of transcription. A ribozyme situated downstream of the gene restored accurate splicing inhibited by the CTD mutant or a mutant poly(A) site, suggesting that cleavage liberates RNA from a niche that is inaccessible to splicing factors. Although ribozyme cleavage enhanced splicing, 3' end processing was impaired, indicating that an intact RNA chain linking the poly(A) site to Pol II is required for optimal processing. Surprisingly, poly(A)(-) beta-globin mRNA with a ribozyme-generated 3' end was exported to the cytoplasm. Ribozyme cleavage can therefore substitute for normal 3' end processing in stimulating splicing and mRNA export. We propose that mRNA biogenesis is coordinated by preventing splicing near the 3' end until the transcript is released by poly(A) site cleavage.


Asunto(s)
Precursores del ARN/metabolismo , Empalme del ARN/fisiología , ARN Catalítico/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética , Línea Celular , ADN/metabolismo , Humanos , Modelos Genéticos , Mutación , Oocitos/metabolismo , Poli A/química , Poli A/genética , Poli A/metabolismo , Procesamiento de Término de ARN 3'/fisiología , ARN Polimerasa II/química , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Empalme del ARN/genética , ARN Mensajero/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...