Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Gene ; 924: 148616, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-38795856

RESUMEN

Transcription initiation is a vital step in the regulation of eukaryotic gene expression. It can be dysregulated in response to various cellular stressors which is associated with numerous human diseases including cancer. Transcription initiation is facilitated via many gene-specific trans-regulatory elements such as transcription factors, activators, and coactivators through their interactions with transcription pre-initiation complex (PIC). These trans-regulatory elements can uniquely facilitate PIC formation (hence, transcription initiation) in response to cellular nutrient stress. Cellular nutrient stress also regulates the activity of other pathways such as target of rapamycin (TOR) pathway. TOR pathway exhibits distinct regulatory mechanisms of transcriptional activation in response to stress. Like TOR pathway, the cell cycle regulatory pathway is also found to be linked to transcriptional regulation in response to cellular stress. Several transcription factors such as p53, C/EBP Homologous Protein (CHOP), activating transcription factor 6 (ATF6α), E2F, transforming growth factor (TGF)-ß, Adenomatous polyposis coli (APC), SMAD, and MYC have been implicated in regulation of transcription of target genes involved in cell cycle progression, apoptosis, and DNA damage repair pathways. Additionally, cellular metabolic and oxidative stressors have been found to regulate the activity of long non-coding RNAs (lncRNA). LncRNA regulates transcription by upregulating or downregulating the transcription regulatory proteins involved in metabolic and cell signaling pathways. Numerous human diseases, triggered by chronic cellular stressors, are associated with abnormal regulation of transcription. Hence, understanding these mechanisms would help unravel the molecular regulatory insights with potential therapeutic interventions. Therefore, here we emphasize the recent advances of regulation of eukaryotic transcription initiation in response to cellular stress.


Asunto(s)
Estrés Fisiológico , Humanos , Estrés Fisiológico/genética , Regulación de la Expresión Génica , Iniciación de la Transcripción Genética , Animales , Transducción de Señal , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/genética
2.
Nucleic Acids Res ; 52(9): 5016-5032, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38471819

RESUMEN

Viruses are master remodelers of the host cell environment in support of infection and virus production. For example, viruses typically regulate cell gene expression through modulating canonical cell promoter activity. Here, we show that Epstein Barr virus (EBV) replication causes 'de novo' transcription initiation at 29674 new transcription start sites throughout the cell genome. De novo transcription initiation is facilitated in part by the unique properties of the viral pre-initiation complex (vPIC) that binds a TATT[T/A]AA, TATA box-like sequence and activates transcription with minimal support by additional transcription factors. Other de novo promoters are driven by the viral transcription factors, Zta and Rta and are influenced by directional proximity to existing canonical cell promoters, a configuration that fosters transcription through existing promoters and transcriptional interference. These studies reveal a new way that viruses interact with the host transcriptome to inhibit host gene expression and they shed light on primal features driving eukaryotic promoter function.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Iniciación de la Transcripción Genética , Replicación Viral , Humanos , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Regiones Promotoras Genéticas , TATA Box , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/virología
3.
Nucleic Acids Res ; 51(16): 8575-8586, 2023 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-37470822

RESUMEN

In addition to being essential for gene expression, transcription is crucial for the maintenance of genome integrity. Here, we undertook a systematic approach, to monitor the assembly kinetics of the pre-initiating RNA Polymerase (Pol) II at promoters at steady state and different stages during recovery from UV irradiation-stress, when pre-initiation and initiation steps have been suggested to be transiently shut down. Taking advantage of the reversible dissociation of pre-initiating Pol II after high salt treatment, we found that de novo recruitment of the available Pol II molecules at active promoters not only persists upon UV at all times tested but occurs significantly faster in the early phase of recovery (2 h) than in unexposed human fibroblasts at the majority of active genes. Our method unveiled groups of genes with significantly different pre-initiation complex (PIC) assembly dynamics after UV that present distinct rates of UV-related mutational signatures in melanoma tumours, providing functional relevance to the importance of keeping transcription initiation active during UV recovery. Our findings uncover novel mechanistic insights further detailing the multilayered transcriptional response to genotoxic stress and link PIC assembly dynamics after exposure to genotoxins with cancer mutational landscapes.


Asunto(s)
ARN Polimerasa II , Iniciación de la Transcripción Genética , Humanos , Daño del ADN , Mutagénesis , Regiones Promotoras Genéticas , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Transcripción Genética , Rayos Ultravioleta , Fibroblastos/metabolismo , Reparación del ADN
5.
Nat Struct Mol Biol ; 28(10): 779-788, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34556871

RESUMEN

Poxviruses express their genes in the cytoplasm of infected cells using a virus-encoded multi-subunit polymerase (vRNAP) and unique transcription factors. We present cryo-EM structures that uncover the complete transcription initiation phase of the poxvirus vaccinia. In the pre-initiation complex, the heterodimeric early transcription factor VETFs/l adopts an arc-like shape spanning the polymerase cleft and anchoring upstream and downstream promoter elements. VETFI emerges as a TBP-like protein that inserts asymmetrically into the DNA major groove, triggers DNA melting, ensures promoter recognition and enforces transcription directionality. The helicase VETFs fosters promoter melting and the phospho-peptide domain (PPD) of vRNAP subunit Rpo30 enables transcription initiation. An unprecedented upstream promoter scrunching mechanism assisted by the helicase NPH-I probably fosters promoter escape and transition into elongation. Our structures shed light on unique mechanisms of poxviral gene expression and aid the understanding of thus far unexplained universal principles in transcription.


Asunto(s)
Iniciación de la Transcripción Genética , Virus Vaccinia/química , Virus Vaccinia/genética , Proteínas Virales/química , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Microscopía por Crioelectrón , ADN Helicasas/química , ADN Helicasas/genética , ADN Viral/química , ADN Viral/metabolismo , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación Viral de la Expresión Génica , Células HeLa , Humanos , Modelos Moleculares , Regiones Promotoras Genéticas , Dominios Proteicos , Subunidades de Proteína , Factores de Transcripción/química , Factores de Transcripción/genética , Proteínas Virales/genética
6.
Elife ; 102021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34137374

RESUMEN

Human bromodomain and extra-terminal domain (BET) family members are promising targets for therapy of cancer and immunoinflammatory diseases, but their mechanisms of action and functional redundancies are poorly understood. Bdf1/2, yeast homologues of the human BET factors, were previously proposed to target transcription factor TFIID to acetylated histone H4, analogous to bromodomains that are present within the largest subunit of metazoan TFIID. We investigated the genome-wide roles of Bdf1/2 and found that their important contributions to transcription extend beyond TFIID function as transcription of many genes is more sensitive to Bdf1/2 than to TFIID depletion. Bdf1/2 co-occupy the majority of yeast promoters and affect preinitiation complex formation through recruitment of TFIID, Mediator, and basal transcription factors to chromatin. Surprisingly, we discovered that hypersensitivity of genes to Bdf1/2 depletion results from combined defects in transcription initiation and early elongation, a striking functional similarity to human BET proteins, most notably Brd4. Our results establish Bdf1/2 as critical for yeast transcription and provide important mechanistic insights into the function of BET proteins in all eukaryotes.


When a healthy cell creates new proteins, it activates a standard two-step biological manufacturing process. Firstly, DNA is transcribed from a specific gene to generate a strand of messenger RNA, or mRNA. Next, this mRNA molecule is translated to create the final protein product. This process of converting DNA into mRNA is supported by a series of helper proteins, including proteins from the bromodomain and extra-terminal domain (BET) family. Cancer cells can become 'addicted' to the process of converting DNA into RNA, leading to the overproduction of mRNA molecules, uncontrolled cell growth and tumor formation. Knocking out BET helper proteins could potentially bring cancer cells under control by halting transcription and preventing tumor growth. However, the precise ways in which BET helper proteins regulate transcription are currently poorly understood, and therefore developing rational ways to target them is a challenge. Building on their previous work, Donczew and Hahn have investigated how two BET helper proteins, Bdf1 and Bdf2, help to regulate transcription in budding yeast. Using a range of genomic techniques, Donczew and Hahn found that Bdf1 and Bdf2 had important roles for initiating transcription and elongating mRNA molecules. Both BET proteins were also involved in recruiting other protein factors to help with the transcription process, including TFIID and Mediator. Based on these findings, it is likely that cooperation between BET proteins, TFIID and Mediator represents a common pathway through which gene expression is regulated across all eukaryotic organisms. Both Bdf1 and Bdf2 were also found to provide the same functions in yeast as similar BET proteins in humans. Using this robust yeast model system to perform further detailed studies of BET factors could therefore provide highly relevant information to expand our understanding of human biology and disease. Ultimately, this research provides important insights into how two members of the BET family of helper proteins contribute to the control of transcription in yeast. This information could be used to guide the design of new drugs for cancer therapy that target not only BET proteins themselves but also other proteins they recruit, including TFIID and Mediator. Such targeted drugs would be expected to be more harmful for cancer cells than for healthy cells, which could reduce unwanted side effects.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Elongación de la Transcripción Genética , Factores de Transcripción , Iniciación de la Transcripción Genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Genoma Fúngico/genética , Humanos , Regiones Promotoras Genéticas/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Proc Natl Acad Sci U S A ; 118(27)2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34187895

RESUMEN

DOT1L, the only H3K79 methyltransferase in human cells and a homolog of the yeast Dot1, normally forms a complex with AF10, AF17, and ENL or AF9, is dysregulated in most cases of mixed-lineage leukemia (MLLr), and has been believed to regulate transcriptional elongation on the basis of its colocalization with RNA polymerase II (Pol II), the sharing of subunits (AF9 and ENL) between the DOT1L and super elongation complexes, and the distribution of H3K79 methylation on both promoters and transcribed regions of active genes. Here we show that DOT1L depletion in erythroleukemic cells reduces its global occupancy without affecting the traveling ratio or the elongation rate (assessed by 4sUDRB-seq) of Pol II, suggesting that DOT1L does not play a major role in elongation in these cells. In contrast, analyses of transcription initiation factor binding reveal that DOT1L and ENL depletions each result in reduced TATA binding protein (TBP) occupancies on thousands of genes. More importantly, DOT1L and ENL depletions concomitantly reduce TBP and Pol II occupancies on a significant fraction of direct (DOT1L-bound) target genes, indicating a role for the DOT1L complex in transcription initiation. Mechanistically, proteomic and biochemical studies suggest that the DOT1L complex may regulate transcriptional initiation by facilitating the recruitment or stabilization of transcription factor IID, likely in a monoubiquitinated H2B (H2Bub1)-enhanced manner. Additional studies show that DOT1L enhances H2Bub1 levels by limiting recruitment of the Spt-Ada-Gcn5-acetyltransferase (SAGA) complex. These results advance our understanding of roles of the DOT1L complex in transcriptional regulation and have important implications for MLLr leukemias.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Leucemia Eritroblástica Aguda/genética , Iniciación de la Transcripción Genética , Línea Celular Tumoral , Cromatina/metabolismo , Regulación Leucémica de la Expresión Génica , Histonas/metabolismo , Humanos , Unión Proteica , ARN Polimerasa II/metabolismo , Elongación de la Transcripción Genética , Factor de Transcripción TFIID/metabolismo , Factores de Elongación Transcripcional/metabolismo , Ubiquitinación
8.
Science ; 372(6546)2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33958484

RESUMEN

The 1.3-megadalton transcription factor IID (TFIID) is required for preinitiation complex (PIC) assembly and RNA polymerase II (Pol II)-mediated transcription initiation on almost all genes. The 26-subunit Mediator stimulates transcription and cyclin-dependent kinase 7 (CDK7)-mediated phosphorylation of the Pol II C-terminal domain (CTD). We determined the structures of human Mediator in the Tail module-extended (at near-atomic resolution) and Tail-bent conformations and structures of TFIID-based PIC-Mediator (76 polypeptides, ~4.1 megadaltons) in four distinct conformations. PIC-Mediator assembly induces concerted reorganization (Head-tilting and Middle-down) of Mediator and creates a Head-Middle sandwich, which stabilizes two CTD segments and brings CTD to CDK7 for phosphorylation; this suggests a CTD-gating mechanism favorable for phosphorylation. The TFIID-based PIC architecture modulates Mediator organization and TFIIH stabilization, underscoring the importance of TFIID in orchestrating PIC-Mediator assembly.


Asunto(s)
Complejo Mediador/química , ARN Polimerasa II/química , Factor de Transcripción TFIID/química , Iniciación de la Transcripción Genética , Microscopía por Crioelectrón , Quinasas Ciclina-Dependientes/química , Quinasas Ciclina-Dependientes/metabolismo , ADN Helicasas/química , Proteínas de Unión al ADN/química , Humanos , Complejo Mediador/metabolismo , Subunidad 1 del Complejo Mediador/química , Modelos Moleculares , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos , Pliegue de Proteína , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , ARN Polimerasa II/metabolismo , Factor de Transcripción TFIID/metabolismo , Factor de Transcripción TFIIH/química , Factor de Transcripción TFIIH/metabolismo , Quinasa Activadora de Quinasas Ciclina-Dependientes
9.
Nature ; 593(7860): 602-606, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33953397

RESUMEN

MicroRNAs (miRNAs) have essential functions during embryonic development, and their dysregulation causes cancer1,2. Altered global miRNA abundance is found in different tissues and tumours, which implies that precise control of miRNA dosage is important1,3,4, but the underlying mechanism(s) of this control remain unknown. The protein complex Microprocessor, which comprises one DROSHA and two DGCR8 proteins, is essential for miRNA biogenesis5-7. Here we identify a developmentally regulated miRNA dosage control mechanism that involves alternative transcription initiation (ATI) of DGCR8. ATI occurs downstream of a stem-loop in DGCR8 mRNA to bypass an autoregulatory feedback loop during mouse embryonic stem (mES) cell differentiation. Deletion of the stem-loop causes imbalanced DGCR8:DROSHA protein stoichiometry that drives irreversible Microprocessor aggregation, reduced primary miRNA processing, decreased mature miRNA abundance, and widespread de-repression of lipid metabolic mRNA targets. Although global miRNA dosage control is not essential for mES cells to exit from pluripotency, its dysregulation alters lipid metabolic pathways and interferes with embryonic development by disrupting germ layer specification in vitro and in vivo. This miRNA dosage control mechanism is conserved in humans. Our results identify a promoter switch that balances Microprocessor autoregulation and aggregation to precisely control global miRNA dosage and govern stem cell fate decisions during early embryonic development.


Asunto(s)
Dosificación de Gen , Estratos Germinativos/metabolismo , MicroARNs/genética , Proteínas de Unión al ARN/genética , Ribonucleasa III/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Células Hep G2 , Humanos , Células K562 , Metabolismo de los Lípidos/genética , Ratones , Regiones Promotoras Genéticas , Iniciación de la Transcripción Genética
10.
Science ; 372(6541)2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33795473

RESUMEN

Transcription factor IID (TFIID) recognizes core promoters and supports preinitiation complex (PIC) assembly for RNA polymerase II (Pol II)-mediated eukaryotic transcription. We determined the structures of human TFIID-based PIC in three stepwise assembly states and revealed two-track PIC assembly: stepwise promoter deposition to Pol II and extensive modular reorganization on track I (on TATA-TFIID-binding element promoters) versus direct promoter deposition on track II (on TATA-only and TATA-less promoters). The two tracks converge at an ~50-subunit holo PIC in identical conformation, whereby TFIID stabilizes PIC organization and supports loading of cyclin-dependent kinase (CDK)-activating kinase (CAK) onto Pol II and CAK-mediated phosphorylation of the Pol II carboxyl-terminal domain. Unexpectedly, TBP of TFIID similarly bends TATA box and TATA-less promoters in PIC. Our study provides structural visualization of stepwise PIC assembly on highly diversified promoters.


Asunto(s)
Complejos Multiproteicos/química , Regiones Promotoras Genéticas , Factor de Transcripción TFIID/química , Iniciación de la Transcripción Genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Hormona Liberadora de Corticotropina/genética , Microscopía por Crioelectrón , Quinasas Ciclina-Dependientes/química , Células HEK293 , Humanos , Fosforilación , Unión Proteica , Dominios Proteicos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , ARN Polimerasa II/química , Porcinos , Urocortinas/genética
11.
Nat Commun ; 12(1): 1072, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33594057

RESUMEN

In addition to nucleosomes, chromatin contains non-histone chromatin-associated proteins, of which the high-mobility group proteins are the most abundant. Chromatin-mediated regulation of transcription involves DNA methylation and histone modifications. However, the order of events and the precise function of high-mobility group proteins during transcription initiation remain unclear. Here we show that high-mobility group AT-hook 2 protein (HMGA2) induces DNA nicks at the transcription start site, which are required by the histone chaperone FACT complex to incorporate nucleosomes containing the histone variant H2A.X. Further, phosphorylation of H2A.X at S139 (γ-H2AX) is required for repair-mediated DNA demethylation and transcription activation. The relevance of these findings is demonstrated within the context of TGFB1 signaling and idiopathic pulmonary fibrosis, suggesting therapies against this lethal disease. Our data support the concept that chromatin opening during transcriptional initiation involves intermediates with DNA breaks that subsequently require DNA repair mechanisms to ensure genome integrity.


Asunto(s)
Desmetilación del ADN , Nucleosomas/metabolismo , Iniciación de la Transcripción Genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Cromatina/química , Cromatina/metabolismo , Células HEK293 , Proteína HMGA2/metabolismo , Histonas/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Ratones , Fosforilación , Fosfoserina/metabolismo , ARN Polimerasa II/metabolismo , Sitio de Iniciación de la Transcripción , Activación Transcripcional/genética , Factor de Crecimiento Transformador beta1/metabolismo
12.
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
13.
J Mol Biol ; 433(14): 166813, 2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-33453189

RESUMEN

The general transcription factor TFIIH contains three ATP-dependent catalytic activities. TFIIH functions in nucleotide excision repair primarily as a DNA helicase and in Pol II transcription initiation as a dsDNA translocase and protein kinase. During initiation, the XPB/Ssl2 subunit of TFIIH couples ATP hydrolysis to dsDNA translocation facilitating promoter opening and the kinase module phosphorylates Pol II to facilitate the transition to elongation. These functions are conserved between metazoans and yeast; however, yeast TFIIH also drives transcription start-site scanning in which Pol II scans downstream DNA to locate productive start-sites. The ten-subunit holo-TFIIH from S. cerevisiae has a processive dsDNA translocase activity required for scanning and a structural role in scanning has been ascribed to the three-subunit TFIIH kinase module. Here, we assess the dsDNA translocase activity of ten-subunit holo- and core-TFIIH complexes (i.e. seven subunits, lacking the kinase module) from both S. cerevisiae and H. sapiens. We find that neither holo nor core human TFIIH exhibit processive translocation, consistent with the lack of start-site scanning in humans. Furthermore, in contrast to holo-TFIIH, the S. cerevisiae core-TFIIH also lacks processive translocation and its dsDNA-stimulated ATPase activity was reduced ~5-fold to a level comparable to the human complexes, potentially explaining the reported upstream shift in start-site observed in vitro in the absence of the S. cerevisiae kinase module. These results suggest that neither human nor S. cerevisiae core-TFIIH can translocate efficiently, and that the S. cerevisiae kinase module functions as a processivity factor to allow for robust transcription start-site scanning.


Asunto(s)
ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN/genética , ADN/metabolismo , Regulación de la Expresión Génica , Factor de Transcripción TFIIH/metabolismo , Sitio de Iniciación de la Transcripción , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Humanos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Iniciación de la Transcripción Genética
14.
Nat Commun ; 11(1): 5560, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33144558

RESUMEN

Cancers result from a set of genetic and epigenetic alterations. Most known oncogenes were identified by gain-of-function mutations in cancer, yet little is known about their epigenetic features. Through integrative analysis of 11,596 epigenomic profiles and mutations from >8200 tumor-normal pairs, we discover broad genic repression domains (BGRD) on chromatin as an epigenetic signature for oncogenes. A BGRD is a widespread enrichment domain of the repressive histone modification H3K27me3 and is further enriched with multiple other repressive marks including H3K9me3, H3K9me2, and H3K27me2. Further, BGRD displays widespread enrichment of repressed cis-regulatory elements. Shortening of BGRDs is linked to derepression of transcription. BGRDs at oncogenes tend to be conserved across normal cell types. Putative tumor-promoting genes and lncRNAs defined using BGRDs are experimentally verified as required for cancer phenotypes. Therefore, BGRDs play key roles in epigenetic regulation of cancer and provide a direction for mutation-independent discovery of oncogenes.


Asunto(s)
Cromatina/genética , Silenciador del Gen , Oncogenes , Neoplasias de la Mama/genética , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Secuencia Conservada/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilación , Motivos de Nucleótidos/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Elongación de la Transcripción Genética , Iniciación de la Transcripción Genética
15.
DNA Repair (Amst) ; 96: 102972, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33007515

RESUMEN

Critical for transcription initiation and bulky lesion DNA repair, TFIIH provides an exemplary system to connect molecular mechanisms to biological outcomes due to its strong genetic links to different specific human diseases. Recent advances in structural and computational biology provide a unique opportunity to re-examine biologically relevant molecular structures and develop possible mechanistic insights for the large dynamic TFIIH complex. TFIIH presents many puzzles involving how its two SF2 helicase family enzymes, XPB and XPD, function in transcription initiation and repair: how do they initiate transcription, detect and verify DNA damage, select the damaged strand for incision, coordinate repair with transcription and cell cycle through Cdk-activating-kinase (CAK) signaling, and result in very different specific human diseases associated with cancer, aging, and development from single missense mutations? By joining analyses of breakthrough cryo-electron microscopy (cryo-EM) structures and advanced computation with data from biochemistry and human genetics, we develop unified concepts and molecular level understanding for TFIIH functions with a focus on structural mechanisms. We provocatively consider that TFIIH may have first evolved from evolutionary pressure for TCR to resolve arrested transcription blocks to DNA replication and later added its key roles in transcription initiation and global DNA repair. We anticipate that this level of mechanistic information will have significant impact on thinking about TFIIH, laying a robust foundation suitable to develop new paradigms for DNA transcription initiation and repair along with insights into disease prevention, susceptibility, diagnosis and interventions.


Asunto(s)
Daño del ADN , Reparación del ADN , Factor de Transcripción TFIIH/metabolismo , Iniciación de la Transcripción Genética , ADN/metabolismo , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Modelos Moleculares , Conformación Proteica , Factor de Transcripción TFIIH/química , Proteína de la Xerodermia Pigmentosa del Grupo D/metabolismo
16.
Nat Commun ; 11(1): 4281, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32855416

RESUMEN

Controlling efficiency and fidelity in the early stage of mitochondrial DNA transcription is crucial for regulating cellular energy metabolism. Conformational transitions of the transcription initiation complex must be central for such control, but how the conformational dynamics progress throughout transcription initiation remains unknown. Here, we use single-molecule fluorescence resonance energy transfer techniques to examine the conformational dynamics of the transcriptional system of yeast mitochondria with single-base resolution. We show that the yeast mitochondrial transcriptional complex dynamically transitions among closed, open, and scrunched states throughout the initiation stage. Then abruptly at position +8, the dynamic states of initiation make a sharp irreversible transition to an unbent conformation with associated promoter release. Remarkably, stalled initiation complexes remain in dynamic scrunching and unscrunching states without dissociating the RNA transcript, implying the existence of backtracking transitions with possible regulatory roles. The dynamic landscape of transcription initiation suggests a kinetically driven regulation of mitochondrial transcription.


Asunto(s)
Mitocondrias/genética , Saccharomyces cerevisiae/genética , Iniciación de la Transcripción Genética , Adenosina Trifosfato , ADN de Hongos/genética , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , ARN de Hongos/genética , ARN de Hongos/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Imagen Individual de Molécula/métodos , Elongación de la Transcripción Genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
ACS Chem Biol ; 15(7): 1765-1772, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32530599

RESUMEN

Dinucleoside polyphosphates (NpnNs) were discovered 50 years ago in all cells. They are often called alarmones, even though the molecular target of the alarm has not yet been identified. Recently, we showed that they serve as noncanonical initiating nucleotides (NCINs) and fulfill the role of 5' RNA caps in Escherichia coli. Here, we present molecular insight into their ability to be used as NCINs by T7 RNA polymerase in the initiation phase of transcription. In general, we observed NpnNs to be equally good substrates as canonical nucleotides for T7 RNA polymerase. Surprisingly, the incorporation of ApnGs boosts the production of RNA 10-fold. This behavior is due to the pairing ability of both purine moieties with the -1 and +1 positions of the antisense DNA strand. Molecular dynamic simulations revealed noncanonical pairing of adenosine with the thymine of the DNA.


Asunto(s)
Fosfatos de Dinucleósidos/genética , ARN/genética , Iniciación de la Transcripción Genética , Bacteriófago T7/enzimología , Emparejamiento Base , ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Fosfatos de Dinucleósidos/metabolismo , Simulación de Dinámica Molecular , Unión Proteica , ARN/metabolismo , Caperuzas de ARN/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
18.
Nat Microbiol ; 5(6): 864-871, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32341479

RESUMEN

Segmented negative-sense RNA viruses (sNSRVs) encode a single-polypeptide polymerase (L protein) or a heterotrimeric polymerase complex to cannibalize host messenger RNA cap structures serving as primers of transcription, and catalyse RNA synthesis. Here, we report the full-length structure of the severe fever with thrombocytopaenia syndrome virus (SFTSV) L protein, as determined by cryogenic electron microscopy at 3.4 Å, leading to an atomic model harbouring three functional parts (an endonuclease, an RNA-dependent RNA polymerase and a cap-binding domain) and two structural domains (an arm domain with a blocker motif and a carboxy-terminal lariat domain). The SFTSV L protein has a compact architecture in which its cap-binding pocket is surprisingly occupied by an Arg finger of the blocker motif, and the endonuclease active centre faces back towards the cap-binding pocket, suggesting that domain rearrangements are necessary to acquire the pre-initiation state of the active site. Our results provide insight into the complete architecture of sNSRV-encoded L protein and further the understanding of sNSRV transcription initiation.


Asunto(s)
Modelos Moleculares , Phlebovirus/genética , Phlebovirus/metabolismo , ARN Viral , Iniciación de la Transcripción Genética , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Secuencia Conservada , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad , Transcripción Genética
19.
Nucleic Acids Res ; 48(1): 157-170, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31722407

RESUMEN

Transcription start sites (TSS) in eukaryotes are characterized by a nucleosome-depleted region (NDR), which appears to be flanked upstream and downstream by strongly positioned nucleosomes incorporating the histone variant H2A.Z. H2A.Z associates with both active and repressed TSS and is important for priming genes for rapid transcriptional activation. However, the determinants of H2A.Z occupancy at specific nucleosomes and its relationship to transcription initiation remain unclear. To further elucidate the specificity of H2A.Z, we determined its genomic localization at single nucleosome resolution, as well as the localization of its chromatin remodelers Swr1 and Ino80. By analyzing H2A.Z occupancy in conjunction with RNA expression data that captures promoter-derived antisense initiation, we find that H2A.Z's bimodal incorporation on either side of the NDR is not a general feature of TSS, but is specifically a marker for bidirectional transcription, such that the upstream flanking -1 H2A.Z-containing nucleosome is more appropriately considered as a +1 H2A.Z nucleosome for antisense transcription. The localization of H2A.Z almost exclusively at the +1 nucleosome suggests that a transcription-initiation dependent process could contribute to its specific incorporation.


Asunto(s)
Adenosina Trifosfatasas/genética , Histonas/genética , ARN Mensajero/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Iniciación de la Transcripción Genética , Adenosina Trifosfatasas/metabolismo , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Histonas/metabolismo , Nucleosomas/química , Nucleosomas/metabolismo , ARN Mensajero/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Sitio de Iniciación de la Transcripción
20.
PLoS Pathog ; 15(6): e1007852, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31188901

RESUMEN

Precise promoter annotation is required for understanding the mechanistic basis of transcription initiation. In the context of complex genomes, such as herpesviruses where there is extensive genic overlap, identification of transcription start sites (TSSs) is particularly problematic and cannot be comprehensively accessed by standard RNA sequencing approaches. Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic gammaherpesvirus and the etiological agent of Kaposi's sarcoma and the B cell lymphoma primary effusion lymphoma (PEL). Here, we leverage RNA annotation and mapping of promoters for analysis of gene expression (RAMPAGE) and define KSHV TSSs transcriptome-wide and at nucleotide resolution in two widely used models of KSHV infection, namely iSLK.219 cells and the PEL cell line TREx-BCBL1-RTA. By mapping TSSs over a 96 h time course of reactivation we confirm 48 of 50 previously identified TSSs. Moreover, we identify over 100 novel transcription start site clusters (TSCs) in each cell line. Our analyses identified cell-type specific differences in TSC positions as well as promoter strength, and defined motifs within viral core promoters. Collectively, by defining TSSs at high resolution we have greatly expanded the transcriptional landscape of the KSHV genome and identified transcriptional control mechanisms at play during KSHV lytic reactivation.


Asunto(s)
Genoma Viral/fisiología , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/fisiología , Iniciación de la Transcripción Genética/fisiología , Activación Viral , Células HEK293 , Infecciones por Herpesviridae/virología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA