Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.195
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Biochem ; 90: 321-348, 2021 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-33770447

RESUMEN

Influenza virus RNA-dependent RNA polymerase (FluPol) transcribes the viral RNA genome in the infected cell nucleus. In the 1970s, researchers showed that viral transcription depends on host RNA polymerase II (RNAP II) activity and subsequently that FluPol snatches capped oligomers from nascent RNAP II transcripts to prime its own transcription. Exactly how this occurs remains elusive. Here, we review recent advances in the mechanistic understanding of FluPol transcription and early events in RNAP II transcription that are relevant to cap-snatching. We describe the known direct interactions between FluPol and the RNAP II C-terminal domain and summarize the transcription-related host factors that have been found to interact with FluPol. We also discuss open questions regarding how FluPol may be targeted to actively transcribing RNAP II and the exact context and timing of cap-snatching, which is presumed to occur after cap completion but before the cap is sequestered by the nuclear cap-binding complex.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Orthomyxoviridae/enzimología , ARN Polimerasa Dependiente del ARN/metabolismo , Transcripción Genética , Proteínas Virales/metabolismo , Humanos , Orthomyxoviridae/patogenicidad , Proteínas de Unión a Caperuzas de ARN/genética , Proteínas de Unión a Caperuzas de ARN/metabolismo , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/genética
2.
Cell ; 184(2): 534-544.e11, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33373586

RESUMEN

Determination of what is the specificity of subunits composing a protein complex is essential when studying gene variants on human pathophysiology. The pore-forming α-subunit KCNQ1, which belongs to the voltage-gated ion channel superfamily, associates to its ß-auxiliary subunit KCNE1 to generate the slow cardiac potassium IKs current, whose dysfunction leads to cardiac arrhythmia. Using pharmacology, gene invalidation, and single-molecule fluorescence assays, we found that KCNE1 fulfils all criteria of a bona fide auxiliary subunit of the TMEM16A chloride channel, which belongs to the anoctamin superfamily. Strikingly, assembly with KCNE1 switches TMEM16A from a calcium-dependent to a voltage-dependent ion channel. Importantly, clinically relevant inherited mutations within the TMEM16A-regulating domain of KCNE1 abolish the TMEM16A modulation, suggesting that the TMEM16A-KCNE1 current may contribute to inherited pathologies. Altogether, these findings challenge the dogma of the specificity of auxiliary subunits regarding protein complexes and questions ion channel classification.


Asunto(s)
Canales de Potasio con Entrada de Voltaje/metabolismo , Subunidades de Proteína/metabolismo , Animales , Anoctamina-1/metabolismo , Calcio/metabolismo , Canales de Cloruro/metabolismo , Células HEK293 , Humanos , Túbulos Renales Proximales/metabolismo , Ratones , Proteínas Mutantes/metabolismo , Péptidos/metabolismo , Polimorfismo Genético , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , Unión Proteica , Dominios Proteicos , Sistema Renina-Angiotensina
3.
Cell ; 175(3): 766-779.e17, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30340042

RESUMEN

The super elongation complex (SEC) is required for robust and productive transcription through release of RNA polymerase II (Pol II) with its P-TEFb module and promoting transcriptional processivity with its ELL2 subunit. Malfunction of SEC contributes to multiple human diseases including cancer. Here, we identify peptidomimetic lead compounds, KL-1 and its structural homolog KL-2, which disrupt the interaction between the SEC scaffolding protein AFF4 and P-TEFb, resulting in impaired release of Pol II from promoter-proximal pause sites and a reduced average rate of processive transcription elongation. SEC is required for induction of heat-shock genes and treating cells with KL-1 and KL-2 attenuates the heat-shock response from Drosophila to human. SEC inhibition downregulates MYC and MYC-dependent transcriptional programs in mammalian cells and delays tumor progression in a mouse xenograft model of MYC-driven cancer, indicating that small-molecule disruptors of SEC could be used for targeted therapy of MYC-induced cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Experimentales/tratamiento farmacológico , Factor B de Elongación Transcripcional Positiva/metabolismo , Proteínas Represoras/metabolismo , Elongación de la Transcripción Genética/efectos de los fármacos , Factores de Elongación Transcripcional/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Drosophila , Femenino , Células HCT116 , Células HEK293 , Respuesta al Choque Térmico , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Polimerasa II/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
4.
Mol Cell ; 84(11): 2053-2069.e9, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38810649

RESUMEN

Facilitates chromatin transcription (FACT) is a histone chaperone that supports transcription through chromatin in vitro, but its functional roles in vivo remain unclear. Here, we analyze the in vivo functions of FACT with the use of multi-omics analysis after rapid FACT depletion from human cells. We show that FACT depletion destabilizes chromatin and leads to transcriptional defects, including defective promoter-proximal pausing and elongation, and increased premature termination of RNA polymerase II. Unexpectedly, our analysis revealed that promoter-proximal pausing depends not only on the negative elongation factor (NELF) but also on the +1 nucleosome, which is maintained by FACT.


Asunto(s)
Cromatina , Proteínas del Grupo de Alta Movilidad , Nucleosomas , Regiones Promotoras Genéticas , ARN Polimerasa II , Transcripción Genética , Factores de Elongación Transcripcional , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Humanos , Factores de Elongación Transcripcional/metabolismo , Factores de Elongación Transcripcional/genética , Cromatina/metabolismo , Cromatina/genética , Nucleosomas/metabolismo , Nucleosomas/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Células HeLa , Ensamble y Desensamble de Cromatina , Células HEK293 , Elongación de la Transcripción Genética , Terminación de la Transcripción Genética
5.
Mol Cell ; 84(7): 1243-1256.e5, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38401543

RESUMEN

Metazoan gene expression regulation involves pausing of RNA polymerase (Pol II) in the promoter-proximal region of genes and is stabilized by DSIF and NELF. Upon depletion of elongation factors, NELF appears to accompany elongating Pol II past pause sites; however, prior work indicates that NELF prevents Pol II elongation. Here, we report cryoelectron microscopy structures of Pol II-DSIF-NELF complexes with NELF in two distinct conformations corresponding to paused and poised states. The paused NELF state supports Pol II stalling, whereas the poised NELF state enables transcription elongation as it does not support a tilted RNA-DNA hybrid. Further, the poised NELF state can accommodate TFIIS binding to Pol II, allowing for Pol II reactivation at paused or backtracking sites. Finally, we observe that the NELF-A tentacle interacts with the RPB2 protrusion and is necessary for pausing. Our results define how NELF can support pausing, reactivation, and elongation by Pol II.


Asunto(s)
Proteínas Nucleares , ARN Polimerasa II , Animales , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Microscopía por Crioelectrón , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
6.
Mol Cell ; 84(17): 3209-3222.e5, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39191261

RESUMEN

RNA polymerases must initiate and pause within a complex chromatin environment, surrounded by nucleosomes and other transcriptional machinery. This environment creates a spatial arrangement along individual chromatin fibers ripe for both competition and coordination, yet these relationships remain largely unknown owing to the inherent limitations of traditional structural and sequencing methodologies. To address this, we employed long-read chromatin fiber sequencing (Fiber-seq) in Drosophila to visualize RNA polymerase (Pol) within its native chromatin context with single-molecule precision along up to 30 kb fibers. We demonstrate that Fiber-seq enables the identification of individual Pol II, nucleosome, and transcription factor footprints, revealing Pol II pausing-driven destabilization of downstream nucleosomes. Furthermore, we demonstrate pervasive direct distance-dependent transcriptional coupling between nearby Pol II genes, Pol III genes, and transcribed enhancers, modulated by local chromatin architecture. Overall, transcription initiation reshapes surrounding nucleosome architecture and couples nearby transcriptional machinery along individual chromatin fibers.


Asunto(s)
Cromatina , Drosophila melanogaster , Nucleosomas , Transcripción Genética , Animales , Nucleosomas/metabolismo , Nucleosomas/genética , Cromatina/metabolismo , Cromatina/genética , Drosophila melanogaster/genética , Drosophila melanogaster/enzimología , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Ensamble y Desensamble de Cromatina , ARN Polimerasa III/metabolismo , ARN Polimerasa III/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/genética
7.
Mol Cell ; 84(13): 2525-2541.e12, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38906142

RESUMEN

The Integrator complex attenuates gene expression via the premature termination of RNA polymerase II (RNAP2) at promoter-proximal pausing sites. It is required for stimulus response, cell differentiation, and neurodevelopment, but how gene-specific and adaptive regulation by Integrator is achieved remains unclear. Here, we identify two sites on human Integrator subunits 13/14 that serve as binding hubs for sequence-specific transcription factors (TFs) and other transcription effector complexes. When Integrator is attached to paused RNAP2, these hubs are positioned upstream of the transcription bubble, consistent with simultaneous TF-promoter tethering. The TFs co-localize with Integrator genome-wide, increase Integrator abundance on target genes, and co-regulate responsive transcriptional programs. For instance, sensory cilia formation induced by glucose starvation depends on Integrator-TF contacts. Our data suggest TF-mediated promoter recruitment of Integrator as a widespread mechanism for targeted transcription regulation.


Asunto(s)
Regulación de la Expresión Génica , Regiones Promotoras Genéticas , ARN Polimerasa II , Factores de Transcripción , Transcripción Genética , Humanos , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Sitios de Unión , Unión Proteica , Células HEK293 , Cilios/metabolismo , Cilios/genética
8.
Mol Cell ; 84(12): 2255-2271.e9, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38851186

RESUMEN

The mechanisms and timescales controlling de novo establishment of chromatin-mediated transcriptional silencing by Polycomb repressive complex 2 (PRC2) are unclear. Here, we investigate PRC2 silencing at Arabidopsis FLOWERING LOCUS C (FLC), known to involve co-transcriptional RNA processing, histone demethylation activity, and PRC2 function, but so far not mechanistically connected. We develop and test a computational model describing proximal polyadenylation/termination mediated by the RNA-binding protein FCA that induces H3K4me1 removal by the histone demethylase FLD. H3K4me1 removal feeds back to reduce RNA polymerase II (RNA Pol II) processivity and thus enhance early termination, thereby repressing productive transcription. The model predicts that this transcription-coupled repression controls the level of transcriptional antagonism to PRC2 action. Thus, the effectiveness of this repression dictates the timescale for establishment of PRC2/H3K27me3 silencing. We experimentally validate these mechanistic model predictions, revealing that co-transcriptional processing sets the level of productive transcription at the locus, which then determines the rate of the ON-to-OFF switch to PRC2 silencing.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Regulación de la Expresión Génica de las Plantas , Silenciador del Gen , Histonas , Proteínas de Dominio MADS , Complejo Represivo Polycomb 2 , ARN Polimerasa II , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Histonas/metabolismo , Histonas/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Complejo Represivo Polycomb 2/metabolismo , Complejo Represivo Polycomb 2/genética , Proteínas de Dominio MADS/genética , Proteínas de Dominio MADS/metabolismo , Transcripción Genética , Poliadenilación , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Terminación de la Transcripción Genética , Cromatina/metabolismo , Cromatina/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética
9.
Mol Cell ; 83(10): 1588-1604.e5, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-37080207

RESUMEN

Gene expression in metazoans is controlled by promoter-proximal pausing of RNA polymerase II, which can undergo productive elongation or promoter-proximal termination. Integrator-PP2A (INTAC) plays a crucial role in determining the fate of paused polymerases, but the underlying mechanisms remain unclear. Here, we establish a rapid degradation system to dissect the functions of INTAC RNA endonuclease and phosphatase modules. We find that both catalytic modules function at most if not all active promoters and enhancers, yet differentially affect polymerase fate. The endonuclease module induces promoter-proximal termination, with its disruption leading to accumulation of elongation-incompetent polymerases and downregulation of highly expressed genes, while elongation-competent polymerases accumulate at lowly expressed genes and non-coding elements, leading to their upregulation. The phosphatase module primarily prevents the release of paused polymerases and limits transcriptional activation, especially for highly paused genes. Thus, both INTAC catalytic modules have unexpectedly general yet distinct roles in dynamic transcriptional control.


Asunto(s)
Monoéster Fosfórico Hidrolasas , ARN Polimerasa II , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Regulación de la Expresión Génica , Activación Transcripcional , Regulación hacia Arriba , Transcripción Genética
10.
Mol Cell ; 83(11): 1798-1809.e7, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37148879

RESUMEN

At active human genes, the +1 nucleosome is located downstream of the RNA polymerase II (RNA Pol II) pre-initiation complex (PIC). However, at inactive genes, the +1 nucleosome is found further upstream, at a promoter-proximal location. Here, we establish a model system to show that a promoter-proximal +1 nucleosome can reduce RNA synthesis in vivo and in vitro, and we analyze its structural basis. We find that the PIC assembles normally when the edge of the +1 nucleosome is located 18 base pairs (bp) downstream of the transcription start site (TSS). However, when the nucleosome edge is located further upstream, only 10 bp downstream of the TSS, the PIC adopts an inhibited state. The transcription factor IIH (TFIIH) shows a closed conformation and its subunit XPB contacts DNA with only one of its two ATPase lobes, inconsistent with DNA opening. These results provide a mechanism for nucleosome-dependent regulation of transcription initiation.


Asunto(s)
Nucleosomas , ARN Polimerasa II , Humanos , Nucleosomas/genética , ARN Polimerasa II/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción TFIIH/metabolismo , ADN/genética , ADN/química , Transcripción Genética , Sitio de Iniciación de la Transcripción
11.
Mol Cell ; 83(24): 4445-4460.e7, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37995689

RESUMEN

The metazoan-specific Integrator complex catalyzes 3' end processing of small nuclear RNAs (snRNAs) and premature termination that attenuates the transcription of many protein-coding genes. Integrator has RNA endonuclease and protein phosphatase activities, but it remains unclear if both are required for complex function. Here, we show IntS6 (Integrator subunit 6) over-expression blocks Integrator function at a subset of Drosophila protein-coding genes, although having no effect on snRNAs or attenuation of other loci. Over-expressed IntS6 titrates protein phosphatase 2A (PP2A) subunits, thereby only affecting gene loci where phosphatase activity is necessary for Integrator function. IntS6 functions analogous to a PP2A regulatory B subunit as over-expression of canonical B subunits, which do not bind Integrator, is also sufficient to inhibit Integrator activity. These results show that the phosphatase module is critical at only a subset of Integrator-regulated genes and point to PP2A recruitment as a tunable step that modulates transcription termination efficiency.


Asunto(s)
Proteínas de Drosophila , Terminación de la Transcripción Genética , Animales , ARN , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , ARN Nuclear Pequeño/genética , Factores de Transcripción/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster
12.
Mol Cell ; 83(2): 186-202.e11, 2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36669479

RESUMEN

PGC-1α is well established as a metazoan transcriptional coactivator of cellular adaptation in response to stress. However, the mechanisms by which PGC-1α activates gene transcription are incompletely understood. Here, we report that PGC-1α serves as a scaffold protein that physically and functionally connects the DNA-binding protein estrogen-related receptor α (ERRα), cap-binding protein 80 (CBP80), and Mediator to overcome promoter-proximal pausing of RNAPII and transcriptionally activate stress-response genes. We show that PGC-1α promotes pausing release in a two-arm mechanism (1) by recruiting the positive transcription elongation factor b (P-TEFb) and (2) by outcompeting the premature transcription termination complex Integrator. Using mice homozygous for five amino acid changes in the CBP80-binding motif (CBM) of PGC-1α that destroy CBM function, we show that efficient differentiation of primary myoblasts to myofibers and timely skeletal muscle regeneration after injury require PGC-1α binding to CBP80. Our findings reveal how PGC-1α activates stress-response gene transcription in a previously unanticipated pre-mRNA quality-control pathway.


Asunto(s)
Precursores del ARN , Factores de Transcripción , Animales , Ratones , Proteínas de Unión al ADN/genética , Músculo Esquelético/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Regiones Promotoras Genéticas , Proteínas de Unión a Caperuzas de ARN/genética , ARN Polimerasa II/metabolismo , Precursores del ARN/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
13.
Mol Cell ; 82(18): 3412-3423.e5, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35973425

RESUMEN

It is unclear how various factors functioning in the transcriptional elongation by RNA polymerase II (RNA Pol II) cooperatively regulate pause/release and productive elongation in living cells. Using an acute protein-depletion approach, we report that SPT6 depletion results in the release of paused RNA Pol II into gene bodies through an impaired recruitment of PAF1C. Short genes demonstrate a release with increased mature transcripts, whereas long genes are released but fail to yield mature transcripts, due to a reduced processivity resulting from both SPT6 and PAF1C loss. Unexpectedly, SPT6 depletion causes an association of NELF with the elongating RNA Pol II on gene bodies, without any observed functional significance on transcriptional elongation pattern, arguing against a role for NELF in keeping RNA Pol II in the paused state. Furthermore, SPT6 depletion impairs heat-shock-induced pausing, pointing to a role for SPT6 in regulating RNA Pol II pause/release through PAF1C recruitment.


Asunto(s)
ARN Polimerasa II , Factores de Transcripción , Respuesta al Choque Térmico , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Factores de Transcripción/genética , Transcripción Genética
14.
Physiol Rev ; 102(2): 993-1024, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486394

RESUMEN

Over the course of more than 500 million years, the kidneys have undergone a remarkable evolution from primitive nephric tubes to intricate filtration-reabsorption systems that maintain homeostasis and remove metabolic end products from the body. The evolutionarily conserved solute carriers organic cation transporter 2 (OCT2) and organic anion transporters 1 and 3 (OAT1/3) coordinate the active secretion of a broad range of endogenous and exogenous substances, many of which accumulate in the blood of patients with kidney failure despite dialysis. Harnessing OCT2 and OAT1/3 through functional preservation or regeneration could alleviate the progression of kidney disease. Additionally, it would improve current in vitro test models that lose their expression in culture. With this review, we explore OCT2 and OAT1/3 regulation from different perspectives: phylogenetic, ontogenetic, and cell dynamic. Our aim is to identify possible molecular targets both to help prevent or compensate for the loss of transport activity in patients with kidney disease and to enable endogenous OCT2 and OAT1/3 induction in vitro in order to develop better models for drug development.


Asunto(s)
Riñón/metabolismo , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Transportador 2 de Cátion Orgánico/metabolismo , Animales , Humanos , Enfermedades Renales/metabolismo , Filogenia
15.
Mol Cell ; 81(5): 983-997.e7, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33539786

RESUMEN

Gene transcription occurs via a cycle of linked events, including initiation, promoter-proximal pausing, and elongation of RNA polymerase II (Pol II). A key question is how transcriptional enhancers influence these events to control gene expression. Here, we present an approach that evaluates the level and change in promoter-proximal transcription (initiation and pausing) in the context of differential gene expression, genome-wide. This combinatorial approach shows that in primary cells, control of gene expression during differentiation is achieved predominantly via changes in transcription initiation rather than via release of Pol II pausing. Using genetically engineered mouse models, deleted for functionally validated enhancers of the α- and ß-globin loci, we confirm that these elements regulate Pol II recruitment and/or initiation to modulate gene expression. Together, our data show that gene expression during differentiation is regulated predominantly at the level of initiation and that enhancers are key effectors of this process.


Asunto(s)
Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , Iniciación de la Transcripción Genética , Globinas alfa/genética , Globinas beta/genética , Animales , Diferenciación Celular , Exones , Feto , Regulación de la Expresión Génica , Biblioteca de Genes , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Intrones , Células K562 , Hígado/citología , Hígado/metabolismo , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Polimerasa II/metabolismo , Transducción de Señal , Globinas alfa/deficiencia , Globinas beta/deficiencia
16.
Mol Cell ; 81(21): 4398-4412.e7, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34520723

RESUMEN

Despite the critical regulatory function of promoter-proximal pausing, the influence of pausing kinetics on transcriptional control remains an active area of investigation. Here, we present Start-TimeLapse-seq (STL-seq), a method that captures the genome-wide kinetics of short, capped RNA turnover and reveals principles of regulation at the pause site. By measuring the rates of release into elongation and premature termination through the inhibition of pause release, we determine that pause-release rates are highly variable, and most promoter-proximal paused RNA polymerase II molecules prematurely terminate (∼80%). The preferred regulatory mechanism upon a hormonal stimulus (20-hydroxyecdysone) is to influence pause-release rather than termination rates. Transcriptional shutdown occurs concurrently with the induction of promoter-proximal termination under hyperosmotic stress, but paused transcripts from TATA box-containing promoters remain stable, demonstrating an important role for cis-acting DNA elements in pausing. STL-seq dissects the kinetics of pause release and termination, providing an opportunity to identify mechanisms of transcriptional regulation.


Asunto(s)
Regulación de la Expresión Génica , Regiones Promotoras Genéticas , ARN Polimerasa II/química , ARN Polimerasa II/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Metilación de ADN , Ecdisterona/química , Perfilación de la Expresión Génica , Técnicas Genéticas , Genoma , Hormonas , Cinética , Mutación , Ósmosis , Unión Proteica , Transducción de Señal
17.
Mol Cell ; 81(17): 3589-3603.e13, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34324863

RESUMEN

Transcription elongation has emerged as a regulatory hub in gene expression of metazoans. A major control point occurs during early elongation before RNA polymerase II (Pol II) is released into productive elongation. Prior research has linked BRD4 with transcription elongation. Here, we use rapid BET protein and BRD4-selective degradation along with quantitative genome-wide approaches to investigate direct functions of BRD4 in Pol II transcription regulation. Notably, as an immediate consequence of acute BRD4 loss, promoter-proximal pause release is impaired, and transcriptionally engaged Pol II past this checkpoint undergoes readthrough transcription. An integrated proteome-wide analysis uncovers elongation and 3'-RNA processing factors as core BRD4 interactors. BRD4 ablation disrupts the recruitment of general 3'-RNA processing factors at the 5'-control region, which correlates with RNA cleavage and termination defects. These studies, performed in human cells, reveal a BRD4-mediated checkpoint and begin to establish a molecular link between 5'-elongation control and 3'-RNA processing.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Nucleares/fisiología , Elongación de la Transcripción Genética/fisiología , Factores de Transcripción/fisiología , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Expresión Génica , Histonas/metabolismo , Humanos , Ratones , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa II/fisiología , Factores de Transcripción/metabolismo , Terminación de la Transcripción Genética/fisiología , Transcripción Genética/genética , Transcripción Genética/fisiología
18.
Genes Dev ; 35(1-2): 147-156, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33303640

RESUMEN

Transcriptionally silent genes must be activated throughout development. This requires nucleosomes be removed from promoters and enhancers to allow transcription factor (TF) binding and recruitment of coactivators and RNA polymerase II (Pol II). Specialized pioneer TFs bind nucleosome-wrapped DNA to perform this chromatin opening by mechanisms that remain incompletely understood. Here, we show that GAGA factor (GAF), a Drosophila pioneer-like factor, functions with both SWI/SNF and ISWI family chromatin remodelers to allow recruitment of Pol II and entry to a promoter-proximal paused state, and also to promote Pol II's transition to productive elongation. We found that GAF interacts with PBAP (SWI/SNF) to open chromatin and allow Pol II to be recruited. Importantly, this activity is not dependent on NURF as previously proposed; however, GAF also synergizes with NURF downstream from this process to ensure efficient Pol II pause release and transition to productive elongation, apparently through its role in precisely positioning the +1 nucleosome. These results demonstrate how a single sequence-specific pioneer TF can synergize with remodelers to activate sets of genes. Furthermore, this behavior of remodelers is consistent with findings in yeast and mice, and likely represents general, conserved mechanisms found throughout eukarya.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Ensamble y Desensamble de Cromatina/genética , Unión Proteica , ARN Polimerasa II/metabolismo , Elongación de la Transcripción Genética
19.
Genes Dev ; 35(9-10): 658-676, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33888562

RESUMEN

The transcription cycle of RNA polymerase II (RNAPII) is governed at multiple points by opposing actions of cyclin-dependent kinases (CDKs) and protein phosphatases, in a process with similarities to the cell division cycle. While important roles of the kinases have been established, phosphatases have emerged more slowly as key players in transcription, and large gaps remain in understanding of their precise functions and targets. Much of the earlier work focused on the roles and regulation of sui generis and often atypical phosphatases-FCP1, Rtr1/RPAP2, and SSU72-with seemingly dedicated functions in RNAPII transcription. Decisive roles in the transcription cycle have now been uncovered for members of the major phosphoprotein phosphatase (PPP) family, including PP1, PP2A, and PP4-abundant enzymes with pleiotropic roles in cellular signaling pathways. These phosphatases appear to act principally at the transitions between transcription cycle phases, ensuring fine control of elongation and termination. Much is still unknown, however, about the division of labor among the PPP family members, and their possible regulation by or of the transcriptional kinases. CDKs active in transcription have recently drawn attention as potential therapeutic targets in cancer and other diseases, raising the prospect that the phosphatases might also present opportunities for new drug development. Here we review the current knowledge and outstanding questions about phosphatases in the context of the RNAPII transcription cycle.


Asunto(s)
Fosfoproteínas Fosfatasas/metabolismo , ARN Polimerasa II/genética , Transcripción Genética/genética , Animales , Sistemas de Liberación de Medicamentos , Humanos , Fosfoproteínas Fosfatasas/genética
20.
Mol Cell ; 78(2): 261-274.e5, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32155413

RESUMEN

RNA polymerase II (RNA Pol II) is generally paused at promoter-proximal regions in most metazoans, and based on in vitro studies, this function has been attributed to the negative elongation factor (NELF). Here, we show that upon rapid depletion of NELF, RNA Pol II fails to be released into gene bodies, stopping instead around the +1 nucleosomal dyad-associated region. The transition to the 2nd pause region is independent of positive transcription elongation factor P-TEFb. During the heat shock response, RNA Pol II is rapidly released from pausing at heat shock-induced genes, while most genes are paused and transcriptionally downregulated. Both of these aspects of the heat shock response remain intact upon NELF loss. We find that NELF depletion results in global loss of cap-binding complex from chromatin without global reduction of nascent transcript 5' cap stability. Thus, our studies implicate NELF functioning in early elongation complexes distinct from RNA Pol II pause-release.


Asunto(s)
Factor B de Elongación Transcripcional Positiva/genética , ARN Polimerasa II/genética , Factores de Transcripción/genética , Transcripción Genética , Animales , Respuesta al Choque Térmico/genética , Humanos , Ratones , Nucleosomas/genética , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA