Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 83(3): 393-403, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36599353

RESUMEN

The positive transcription elongation factor b (P-TEFb) is composed of cyclins T1 or T2 and cyclin-dependent kinase 9 that regulate the elongation phase of transcription by RNA polymerase II. By antagonizing negative elongation factors and phosphorylating the C-terminal domain of RNA polymerase II, P-TEFb facilitates the elongation and co-transcriptional processing of nascent transcripts. This step is critical for the expression of most eukaryotic genes. In growing cells, P-TEFb is regulated negatively by its reversible associations with HEXIM1/2 in the 7SK snRNP and positively by a number of transcription factors, as well as the super elongation complex. In resting cells, P-TEFb falls apart, and cyclin T1 is degraded by the proteasome. This complex regulation of P-TEFb has evolved for the precise temporal and spatial regulation of gene expression in the organism. Its dysregulation contributes to inflammatory and neoplastic conditions.


Asunto(s)
Factor B de Elongación Transcripcional Positiva , ARN Polimerasa II , Humanos , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Células HeLa , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Ciclina T/genética , Ciclina T/metabolismo , Transcripción Genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Mol Cell ; 62(1): 34-46, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27058786

RESUMEN

Studying cancer metabolism gives insight into tumorigenic survival mechanisms and susceptibilities. In melanoma, we identify HEXIM1, a transcription elongation regulator, as a melanoma tumor suppressor that responds to nucleotide stress. HEXIM1 expression is low in melanoma. Its overexpression in a zebrafish melanoma model suppresses cancer formation, while its inactivation accelerates tumor onset in vivo. Knockdown of HEXIM1 rescues zebrafish neural crest defects and human melanoma proliferation defects that arise from nucleotide depletion. Under nucleotide stress, HEXIM1 is induced to form an inhibitory complex with P-TEFb, the kinase that initiates transcription elongation, to inhibit elongation at tumorigenic genes. The resulting alteration in gene expression also causes anti-tumorigenic RNAs to bind to and be stabilized by HEXIM1. HEXIM1 plays an important role in inhibiting cancer cell-specific gene transcription while also facilitating anti-cancer gene expression. Our study reveals an important role for HEXIM1 in coupling nucleotide metabolism with transcriptional regulation in melanoma.


Asunto(s)
Melanoma/metabolismo , Factor B de Elongación Transcripcional Positiva/genética , Pirimidinas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/patología , Melanoma Experimental , Proteínas Oncogénicas/genética , Factores de Transcripción , Transcripción Genética , Proteínas Supresoras de Tumor/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
Nucleic Acids Res ; 50(9): 5000-5013, 2022 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-35524561

RESUMEN

P-TEFb, composed of CycT1 and CDK9, regulates the elongation of transcription by RNA polymerase II. In proliferating cells, it is regulated by 7SK snRNA in the 7SK snRNP complex. In resting cells, P-TEFb is absent, because CycT1 is dephosphorylated, released from CDK9 and rapidly degraded. In this study, we identified the mechanism of this degradation. We mapped the ubiquitination and degradation of free CycT1 to its N-terminal region from positions 1 to 280. This region is ubiquitinated at six lysines, where E3 ligases Siah1 and Siah2 bind and degrade these sequences. Importantly, the inhibition of Siah1/2 rescued the expression of free CycT1 in proliferating as well as resting primary cells. We conclude that Siah1/2 are the E3 ligases that bind and degrade the dissociated CycT1 in resting, terminally differentiated, anergic and/or exhausted cells.


Asunto(s)
Factor B de Elongación Transcripcional Positiva , Factores de Transcripción , Ubiquitina-Proteína Ligasas/metabolismo , Fenómenos Fisiológicos Celulares , Ciclina T/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , ARN Polimerasa II/metabolismo , ARN Nuclear Pequeño , Factores de Transcripción/metabolismo , Transcripción Genética
4.
PLoS Pathog ; 14(11): e1007402, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30395647

RESUMEN

Transcription of HIV provirus is a key step of the viral cycle, and depends on the recruitment of the cellular positive transcription elongation factor b (P-TEFb) to the HIV promoter. The viral transactivator Tat can displace P-TEFb from the 7SK small nuclear ribonucleoprotein, where it is bound and inactivated by HEXIM1, and bring it to TAR, which allows the stalled RNA polymerase II to transition to successful transcription elongation. In this study, we designed a chimeric inhibitor of HIV transcription by combining functional domains from HEXIM1 and Tat. The chimera (HT1) potently inhibited gene expression from the HIV promoter, by competing with Tat for TAR and P-TEFb binding, while keeping the latter inactive. HT1 inhibited spreading infection as well as viral reactivation in lymphocyte T cell line models of HIV latency, with little effect on cellular transcription and metabolism. This proof-of-concept study validates an innovative approach to interfering with HIV transcription via peptide mimicry and competition for RNA-protein interactions. HT1 represents a new candidate for HIV therapy, or HIV cure via the proposed block and lock strategy.


Asunto(s)
Infecciones por VIH/terapia , VIH-1/fisiología , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Replicación Viral/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Células HEK293 , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Duplicado del Terminal Largo de VIH , Seropositividad para VIH , VIH-1/genética , VIH-1/metabolismo , Células HeLa , Humanos , Células Jurkat , Provirus/genética , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Recombinantes de Fusión/genética , Factores de Transcripción , Latencia del Virus , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
5.
PLoS Pathog ; 14(1): e1006830, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29304101

RESUMEN

The lentiviral protein Viral Infectivity Factor (Vif) counteracts the antiviral effects of host APOBEC3 (A3) proteins and contributes to persistent HIV infection. Vif targets A3 restriction factors for ubiquitination and proteasomal degradation by recruiting them to a multi-protein ubiquitin E3 ligase complex. Here, we describe a degradation-independent mechanism of Vif-mediated antagonism that was revealed through detailed structure-function studies of antibody antigen-binding fragments (Fabs) to the Vif complex. Two Fabs were found to inhibit Vif-mediated A3 neutralization through distinct mechanisms: shielding A3 from ubiquitin transfer and blocking Vif E3 assembly. Combined biochemical, cell biological and structural studies reveal that disruption of Vif E3 assembly inhibited A3 ubiquitination but was not sufficient to restore its packaging into viral particles and antiviral activity. These observations establish that Vif can neutralize A3 family members in a degradation-independent manner. Additionally, this work highlights the potential of Fabs as functional probes, and illuminates how Vif uses a multi-pronged approach involving both degradation dependent and independent mechanisms to suppress A3 innate immunity.


Asunto(s)
Antivirales/farmacología , Citosina Desaminasa/metabolismo , Fragmentos Fab de Inmunoglobulinas/química , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/inmunología , Desaminasas APOBEC , Antivirales/química , Proteínas Cullin/química , Proteínas Cullin/metabolismo , Citidina Desaminasa , Células HEK293 , Infecciones por VIH/inmunología , Infecciones por VIH/terapia , Infecciones por VIH/virología , VIH-1/inmunología , VIH-1/metabolismo , Humanos , Ubiquitina/metabolismo , Ubiquitinación , Ensamble de Virus , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/química
6.
J Biol Chem ; 293(14): 4993-5004, 2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463681

RESUMEN

Autoimmune regulator (AIRE) and nuclear factor-κB (NF-κB) are transcription factors (TFs) that direct the expression of individual genes and gene clusters. Bromodomain-containing protein 4 (BRD4) is an epigenetic regulator that recognizes and binds to acetylated histones. BRD4 also has been reported to promote interactions between the positive transcription elongation factor b (P-TEFb) and AIRE or P-TEFb and NF-κB subunit p65. Here, we report that AIRE and p65 bind to P-TEFb independently of BRD4. JQ1, a compound that disrupts interactions between BRD4 and acetylated proteins, does not decrease transcriptional activities of AIRE or p65. Moreover, siRNA-mediated inactivation of BRD4 alone or in combination with JQ1 had no effects on AIRE- and NF-κB-targeted genes on plasmids and in chromatin and on interactions between P-TEFb and AIRE or NF-κB. Finally, ChIP experiments revealed that recruitment of P-TEFb to AIRE or p65 to transcription complexes was independent of BRD4. We conclude that direct interactions between AIRE, NF-κB, and P-TEFb result in efficient transcription of their target genes.


Asunto(s)
Proteínas Nucleares/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteínas de Ciclo Celular , Eliminación de Gen , Células HEK293 , Humanos , Insulina/genética , Proteínas Nucleares/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Regiones Promotoras Genéticas , Mapas de Interacción de Proteínas , Interferencia de ARN , Factores de Transcripción/genética , Proteína AIRE
7.
Genes Dev ; 25(20): 2158-72, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22012619

RESUMEN

Various cyclin-dependent kinase (Cdk) complexes have been implicated in the regulation of transcription. In this study, we identified a 70-kDa Cyclin K (CycK) that binds Cdk12 and Cdk13 to form two different complexes (CycK/Cdk12 or CycK/Cdk13) in human cells. The CycK/Cdk12 complex regulates phosphorylation of Ser2 in the C-terminal domain of RNA polymerase II and expression of a small subset of human genes, as revealed in expression microarrays. Depletion of CycK/Cdk12 results in decreased expression of predominantly long genes with high numbers of exons. The most prominent group of down-regulated genes are the DNA damage response genes, including the critical regulators of genomic stability: BRCA1 (breast and ovarian cancer type 1 susceptibility protein 1), ATR (ataxia telangiectasia and Rad3-related), FANCI, and FANCD2. We show that CycK/Cdk12, rather than CycK/Cdk13, is necessary for their expression. Nuclear run-on assays and chromatin immunoprecipitations with RNA polymerase II on the BRCA1 and FANCI genes suggest a transcriptional defect in the absence of CycK/Cdk12. Consistent with these findings, cells without CycK/Cdk12 induce spontaneous DNA damage and are sensitive to a variety of DNA damage agents. We conclude that through regulation of expression of DNA damage response genes, CycK/Cdk12 protects cells from genomic instability. The essential role of CycK for organisms in vivo is further supported by the result that genetic inactivation of CycK in mice causes early embryonic lethality.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Daño del ADN/genética , Regulación del Desarrollo de la Expresión Génica , Inestabilidad Genómica , Animales , Proteína Quinasa CDC2/metabolismo , Quinasa 9 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/genética , Ciclinas/genética , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Noqueados , Fosforilación
8.
J Virol ; 91(11)2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28331090

RESUMEN

P-element-induced wimpy-like (Piwil) proteins restrict the replication of mobile genetic elements in the germ line. They are also expressed in many transformed cell lines. In this study, we discovered that the human Piwil 2 (Hili) protein can also inhibit HIV replication, especially in activated CD4+ T cells that are the preferred target cells for this virus in the infected host. Although resting cells did not express Hili, its expression was rapidly induced following T cell activation. In these cells and transformed cell lines, depletion of Hili increased levels of viral proteins and new viral particles. Further studies revealed that Hili binds to tRNA. Some of the tRNAs represent rare tRNA species, whose codons are overrepresented in the viral genome. Targeting tRNAArg(UCU) with an antisense oligonucleotide replicated effects of Hili and also inhibited HIV replication. Finally, Hili also inhibited the retrotransposition of the endogenous intracysternal A particle (IAP) by a similar mechanism. Thus, Hili joins a list of host proteins that inhibit the replication of HIV and other mobile genetic elements.IMPORTANCE Piwil proteins inhibit the movement of mobile genetic elements in the germ line. In their absence, sperm does not form and male mice are sterile. This inhibition is thought to occur via small Piwi-interacting RNAs (piRNAs). However, in some species and in human somatic cells, Piwil proteins bind primarily to tRNA. In this report, we demonstrate that human Piwil proteins, especially Hili, not only bind to select tRNA species, including rare tRNAs, but also inhibit HIV replication. Importantly, T cell activation induces the expression of Hili in CD4+ T cells. Since Hili also inhibited the movement of an endogenous retrovirus (IAP), our finding shed new light on this intracellular resistance to exogenous and endogenous retroviruses as well as other mobile genetic elements.


Asunto(s)
Proteínas Argonautas/metabolismo , VIH-1/fisiología , Activación de Linfocitos , Replicación Viral , Proteínas Argonautas/deficiencia , Proteínas Argonautas/genética , Proteínas Argonautas/inmunología , Línea Celular , Retrovirus Endógenos/metabolismo , Células HEK293 , VIH-1/genética , Células HeLa , Humanos , Oligonucleótidos Antisentido/genética , Unión Proteica , ARN Interferente Pequeño/metabolismo , ARN de Transferencia/metabolismo , ARN de Transferencia de Arginina/genética , ARN de Transferencia de Arginina/metabolismo , Linfocitos T/virología
9.
J Biol Chem ; 291(34): 17953-63, 2016 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-27365398

RESUMEN

The autoimmune regulator (AIRE) is a transcription factor which is expressed in medullary thymic epithelial cells. It directs the expression of otherwise tissue-specific antigens, which leads to the elimination of autoreactive T cells during development. AIRE is modified post-translationally by phosphorylation and ubiquitylation. In this report we connected these modifications. AIRE, which is phosphorylated on two specific residues near its N terminus, then binds to the F-box protein 3 (FBXO3) E3 ubiquitin ligase. In turn, this SCF(FBXO3) (SKP1-CUL1-F box) complex ubiquitylates AIRE, increases its binding to the positive transcription elongation factor b (P-TEFb), and potentiates its transcriptional activity. Because P-TEFb is required for the transition from initiation to elongation of transcription, this interaction ensures proper expression of AIRE-responsive tissue-specific antigens in the thymus.


Asunto(s)
Proteínas F-Box/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología , Ubiquitinación/fisiología , Animales , Proteínas F-Box/genética , Células HEK293 , Humanos , Ratones , Fosforilación/fisiología , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Dominios Proteicos , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Factores de Transcripción/genética , Proteína AIRE
10.
PLoS Pathog ; 11(7): e1005063, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26225566

RESUMEN

The persistence of latently infected cells in patients under combinatory antiretroviral therapy (cART) is a major hurdle to HIV-1 eradication. Strategies to purge these reservoirs are needed and activation of viral gene expression in latently infected cells is one promising strategy. Bromodomain and Extraterminal (BET) bromodomain inhibitors (BETi) are compounds able to reactivate latent proviruses in a positive transcription elongation factor b (P-TEFb)-dependent manner. In this study, we tested the reactivation potential of protein kinase C (PKC) agonists (prostratin, bryostatin-1 and ingenol-B), which are known to activate NF-κB signaling pathway as well as P-TEFb, used alone or in combination with P-TEFb-releasing agents (HMBA and BETi (JQ1, I-BET, I-BET151)). Using in vitro HIV-1 post-integration latency model cell lines of T-lymphoid and myeloid lineages, we demonstrated that PKC agonists and P-TEFb-releasing agents alone acted as potent latency-reversing agents (LRAs) and that their combinations led to synergistic activation of HIV-1 expression at the viral mRNA and protein levels. Mechanistically, combined treatments led to higher activations of P-TEFb and NF-κB than the corresponding individual drug treatments. Importantly, we observed in ex vivo cultures of CD8+-depleted PBMCs from 35 cART-treated HIV-1+ aviremic patients that the percentage of reactivated cultures following combinatory bryostatin-1+JQ1 treatment was identical to the percentage observed with anti-CD3+anti-CD28 antibodies positive control stimulation. Remarkably, in ex vivo cultures of resting CD4+ T cells isolated from 15 HIV-1+ cART-treated aviremic patients, the combinations bryostatin-1+JQ1 and ingenol-B+JQ1 released infectious viruses to levels similar to that obtained with the positive control stimulation. The potent effects of these two combination treatments were already detected 24 hours post-stimulation. These results constitute the first demonstration of LRA combinations exhibiting such a potent effect and represent a proof-of-concept for the co-administration of two different types of LRAs as a potential strategy to reduce the size of the latent HIV-1 reservoirs.


Asunto(s)
Brioestatinas/farmacología , Linfocitos T CD4-Positivos/virología , Regulación Viral de la Expresión Génica/efectos de los fármacos , VIH-1/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de los fármacos , Diterpenos/metabolismo , VIH-1/fisiología , Humanos , Factor B de Elongación Transcripcional Positiva/metabolismo , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos
11.
J Biol Chem ; 290(3): 1829-36, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25492871

RESUMEN

Regulation of transcription elongation by positive transcription elongation factor b (P-TEFb) plays a central role in determining the state of cell activation, proliferation, and differentiation. In cells, P-TEFb exists in active and inactive forms. Its release from the inactive 7SK small nuclear ribonucleoprotein complex is a critical step for P-TEFb to activate transcription elongation. However, no good method exists to analyze this P-TEFb equilibrium in living cells. Only inaccurate and labor-intensive cell-free biochemical assays are currently available. In this study, we present the first experimental system to monitor P-TEFb activation in living cells. We created a bimolecular fluorescence complementation assay to detect interactions between P-TEFb and its substrate, the C-terminal domain of RNA polymerase II. When cells were treated with suberoylanilide hydroxamic acid, which releases P-TEFb from the 7SK small nuclear ribonucleoprotein, they turned green. Other known P-TEFb-releasing agents, including histone deacetylase inhibitors, bromodomain and extraterminal bromodomain inhibitors, and protein kinase C agonists, also scored positive in this assay. Finally, we identified 5'-azacytidine as a new P-TEFb-releasing agent. This release of P-TEFb correlated directly with activation of human HIV and HEXIM1 transcription. Thus, our visualization of P-TEFb activation by fluorescent complementation assay could be used to find new P-TEFb-releasing agents, compare different classes of agents, and assess their efficacy singly and/or in combination.


Asunto(s)
Factor B de Elongación Transcripcional Positiva/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Apoptosis , Azacitidina/química , Proteínas Bacterianas/química , Sistema Libre de Células , Prueba de Complementación Genética , Glicerol/química , Células HEK293 , Células HeLa , Inhibidores de Histona Desacetilasas/química , Humanos , Ácidos Hidroxámicos/química , Proteínas Luminiscentes/química , Microscopía Fluorescente , Plásmidos/metabolismo , Proteína Quinasa C/metabolismo , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/metabolismo , Factores de Tiempo , Transcripción Genética , Vorinostat
12.
Mol Cell ; 29(5): 539-40, 2008 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-18342601

RESUMEN

In a recent issue of Cell Host & Microbe, Kwon et al. (2008) report that the human immunodeficiency virus (HIV) transactivator Tat inhibits the SIRT1 deacetylase, resulting in increased acetylation of the NF-kappaB p65 subunit and subsequently in T cell hyperactivation.


Asunto(s)
Sirtuinas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Síndrome de Inmunodeficiencia Adquirida/inmunología , Animales , VIH-1/genética , VIH-1/metabolismo , Humanos , Activación de Linfocitos , Ratones , FN-kappa B/metabolismo , Sirtuina 1 , Sirtuinas/genética , Linfocitos T/inmunología , Transcripción Genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
13.
J Biol Chem ; 289(30): 21181-90, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24917669

RESUMEN

The positive transcription elongation factor b (P-TEFb), comprised of cyclin-dependent kinase 9 (CDK9) and cyclins T1 (CycT1) or T2 (CycT2), activates eukaryotic transcription elongation. In growing cells, P-TEFb exists in active and inactive forms. In the latter, it is incorporated into the 7SK small nuclear ribonucleoprotein, which contains hexamethylene bisacetamide-induced proteins (HEXIM) 1 or 2, La-related protein 7 (LaRP7), methyl phosphate capping enzyme, and 7SK small nuclear RNA (7SK). HEXIM1 inhibits the kinase activity of CDK9 via interactions between 7SK, HEXIM1, and CycT1. LaRP7 and methyl phosphate capping enzyme interact with 7SK independently of HEXIM1 and P-TEFb. To analyze genetic interactions between HEXIM1 and/or LaRP7 and 7SK using a cell-based system, we established artificial heterologous RNA tethering assays in which reporter gene expression depended on interactions between selected regions of 7SK and its cognate binding partners fused to a strong activator. This system enabled us to map the HEXIM1- and LaRP7- binding regions of 7SK. Assays with various mutant 7SK plasmid targets revealed that the 5'U-Ubulge and central loop of stem-loop I or RNA motif 3 of 7SK are required for transactivation, suggesting that HEXIM1 and CycT1 form a combinatorial binding surface for 7SK. Moreover, a region in HEXIM1 C-terminal to its previously mapped RNA-binding motif was also required for interactions between HEXIM1 and 7SK. Finally, a tyrosine-to-alanine mutation in HEXIM1, which is critical for its inhibitory effect on CDK9, changed HEXIM1 into an activator. These cell-based assays elucidate this important aspect of transcription elongation in vivo.


Asunto(s)
Ribonucleoproteínas Nucleares Pequeñas/genética , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Secuencias de Aminoácidos , Ciclina T/genética , Ciclina T/metabolismo , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Células HEK293 , Células HeLa , Humanos , Conformación de Ácido Nucleico , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo , Relación Estructura-Actividad , Factores de Transcripción , Activación Transcripcional/genética
14.
J Biol Chem ; 289(14): 9918-25, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24515107

RESUMEN

By phosphorylating negative elongation factors and the C-terminal domain of RNA polymerase II (RNAPII), positive transcription elongation factor b (P-TEFb), which is composed of CycT1 or CycT2 and CDK9, activates eukaryotic transcription elongation. In growing cells, it is found in active and inactive forms. In the former, free P-TEFb is a potent transcriptional coactivator. In the latter, it is inhibited by HEXIM1 or HEXIM2 in the 7SK small nuclear ribonucleoprotein (snRNP), which contains, additionally, 7SK snRNA, methyl phosphate-capping enzyme (MePCE), and La-related protein 7 (LARP7). This P-TEFb equilibrium determines the state of growth and proliferation of the cell. In this study, the release of P-TEFb from the 7SK snRNP led to increased synthesis of HEXIM1 but not HEXIM2 in HeLa cells, and this occurred only from an unannotated, proximal promoter. ChIP with sequencing revealed P-TEFb-sensitive poised RNA polymerase II at this proximal but not the previously annotated distal HEXIM1 promoter. Its immediate upstream sequences were fused to luciferase reporters and were found to be responsive to many P-TEFb-releasing compounds. The superelongation complex subunits AF4/FMR2 family member 4 (AFF4) and elongation factor RNA polymerase II 2 (ELL2) were recruited to this proximal promoter after P-TEFb release and were required for its transcriptional effects. Thus, P-TEFb regulates its own equilibrium in cells, most likely to maintain optimal cellular homeostasis.


Asunto(s)
Ciclina T/metabolismo , Quinasa 9 Dependiente de la Ciclina/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Transcripción Genética/fisiología , Ciclina T/genética , Quinasa 9 Dependiente de la Ciclina/genética , Células HEK293 , Células HeLa , Humanos , Metiltransferasas/biosíntesis , Metiltransferasas/genética , Factor B de Elongación Transcripcional Positiva/genética , Biosíntesis de Proteínas/fisiología , Proteínas de Unión al ARN/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Ribonucleoproteínas/biosíntesis , Ribonucleoproteínas/genética , Ribonucleoproteínas Nucleares Pequeñas/biosíntesis , Ribonucleoproteínas Nucleares Pequeñas/genética , Factores de Transcripción , Factores de Elongación Transcripcional
16.
Nat Rev Immunol ; 3(2): 97-107, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12563294

RESUMEN

Viruses that induce chronic infections can evade immune responses. HIV is a prototype of this class of pathogen. Not only does it mutate rapidly and make its surface components difficult to access by neutralizing antibodies, but it also creates cellular hideouts, establishes proviral latency, removes cell-surface receptors and destroys immune effectors to escape eradication. A better understanding of these strategies might lead to new approaches in the fight against AIDS.


Asunto(s)
Infecciones por VIH/inmunología , Apoptosis , Genoma Viral , VIH/genética , VIH/inmunología , VIH/patogenicidad , VIH/fisiología , Infecciones por VIH/virología , Humanos , Modelos Inmunológicos , Provirus/inmunología , Linfocitos T/inmunología , Linfocitos T/virología , Replicación Viral
17.
J Biol Chem ; 288(20): 14400-14407, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23539624

RESUMEN

Numerous studies have looked at the effects of histone deacetylase inhibitors (HDACis) on HIV reactivation in established transformed cell lines and primary CD4(+) T cells. However, their findings remain confusing, and differences between effects of class I- and class II-specific HDACis persist. Because no clear picture emerged, we decided to determine how HDACis reactivate HIV in transformed cell lines and primary cells. We found that neither histone H3 nor tubulin acetylation correlated with HIV reactivation in Jurkat and HeLa cells. Rather, HDACis that could reactivate HIV in chromatin or on episomal plasmids also released free positive transcription elongation factor b (P-TEFb) from its inhibitory 7SK snRNP. In resting primary CD4(+) T cells, where levels of P-TEFb are vanishingly low, the most potent HDACi, suberoylanilide hydroxyamic acid (SAHA), had minimal effects. In contrast, when these cells were treated with a PKC agonist, bryostatin 1, which increased levels of P-TEFb, then SAHA once again reactivated HIV. We conclude that HDACis, which can reactivate HIV, work via the release of free P-TEFb from the 7SK snRNP.


Asunto(s)
Infecciones por VIH/virología , VIH-1/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Factor B de Elongación Transcripcional Positiva/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Linfocitos T CD4-Positivos/citología , Línea Celular , Regulación Viral de la Expresión Génica , Infecciones por VIH/tratamiento farmacológico , Células HeLa , Histonas/química , Histonas/metabolismo , Humanos , Células Jurkat , Factor B de Elongación Transcripcional Positiva/genética , Factores de Transcripción , Tubulina (Proteína)/metabolismo
18.
Nucleic Acids Res ; 40(18): 9160-70, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22821562

RESUMEN

The positive transcription elongation factor b (P-TEFb) regulates RNA polymerase II elongation. In cells, P-TEFb partitions between small active and larger inactive states. In the latter, HEXIM1 binds to 7SK snRNA and recruits as well as inactivates P-TEFb in the 7SK snRNP. Several stimuli can affect this P-TEFb equilibrium. In this study, we demonstrate that protein kinase C (PKC) phosphorylates the serine at position158 (S158) in HEXIM1. This phosphorylated HEXIM1 protein neither binds to 7SK snRNA nor inhibits P-TEFb. Phorbol esters or the engagement of the T cell antigen receptor, which activate PKC and the expression of the constitutively active (CA) PKCθ protein, which is found in T cells, inhibit the formation of the 7SK snRNP. All these stimuli increase P-TEFb-dependent transcription. In contrast, the kinase-negative PKCθ and the mutant HEXIM1 (S158A) proteins block effects of these PKC-activating stimuli. These results indicate that the phosphorylation of HEXIM1 by PKC represents a major regulatory step of P-TEFb activity in cells.


Asunto(s)
Factor B de Elongación Transcripcional Positiva/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteínas de Unión al ARN/metabolismo , Secuencia de Aminoácidos , Línea Celular , Humanos , Células Jurkat , Datos de Secuencia Molecular , FN-kappa B/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , ARN Nuclear Pequeño/metabolismo , Proteínas de Unión al ARN/química , Receptores de Antígenos de Linfocitos T/inmunología , Serina/metabolismo , Factores de Transcripción
19.
J Biol Chem ; 287(43): 36609-16, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22952229

RESUMEN

By phosphorylating elongation factors and the C-terminal domain of RNA polymerase II, the positive transcription elongation factor b (P-TEFb) is the critical kinase for transcription elongation and co-transcriptional processing of eukaryotic genes. It exists in inactive small nuclear ribonucleoprotein (7SK snRNP) and active (free P-TEFb) complexes in cells. The P-TEFb equilibrium determines the state of cellular activation, proliferation, and differentiation. Free P-TEFb, which is required for growth, can be recruited to RNA polymerase II via transcription factors, BRD4, or the super elongation complex (SEC). UV light, various signaling cascades, transcriptional blockade, or compounds such as hexamethylene bisacetamide (HMBA), suberoylanilide hydroxamic acid (SAHA), and other histone deacetylase inhibitors lead to a rapid release of free P-TEFb, followed by its reassembly into the 7SK snRNP. As a consequence, transcription of HEXIM1, a critical 7SK snRNP subunit, and HIV is induced. In this study, we found that a bromodomain and extra-terminal (BET) bromodomain inhibitor, JQ1, which inhibits BRD4 by blocking its association with chromatin, also leads to the rapid release of free P-TEFb from the 7SK snRNP. Indeed, JQ1 transiently increased levels of free P-TEFb and BRD4·P-TEFb and SEC·P-TEFb complexes in cells. As a consequence, the levels of HEXIM1 and HIV proteins rose. Importantly, the knockdown of ELL2, a subunit of the SEC, blocked the ability of JQ1 to increase HIV transcription. Finally, the effects of JQ1 and HMBA or SAHA on the P-TEFb equilibrium were cooperative. We conclude that HMBA, SAHA, and JQ1 affect transcription elongation by a similar and convergent mechanism.


Asunto(s)
Proteínas Nucleares/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , ARN Polimerasa II/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Elongación de la Transcripción Genética , Factores de Transcripción/metabolismo , Acetamidas/farmacología , Azepinas/farmacología , Proteínas de Ciclo Celular , Línea Celular , Técnicas de Silenciamiento del Gen , VIH-1/genética , VIH-1/metabolismo , Humanos , Proteínas Nucleares/genética , Factor B de Elongación Transcripcional Positiva/genética , ARN Polimerasa II/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/genética , Factores de Transcripción/genética , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo , Triazoles/farmacología , Rayos Ultravioleta , Proteínas Virales/genética , Proteínas Virales/metabolismo
20.
J Virol ; 86(19): 10517-23, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22811528

RESUMEN

Moloney leukemia virus type 10 protein (MOV10) is an RNA helicase that is induced by type I interferon. It inhibits HIV replication at several steps of its replicative cycle. Of interest, MOV10 is a component of mRNA processing (P) bodies, which inhibit retrotransposition (RTP) of intracisternal A particles (IAP). In this report, we studied the effects of MOV10 on IAP RTP and its dependence on P bodies. Indeed, MOV10 inhibited IAP RTP. It decreased significantly not only the products of reverse transcriptase but also its endogenous activity. MOV10 also associated with IAP RNA. Furthermore, although it was found in IAP virus-like particles, it did not affect their incorporation of IAP RNA, primer tRNAPhe (phenylalanine tRNA), or IAP Gag. Concerning P bodies, the exogenously expressed MOV10 had no effect on their size and number, and the inhibition of IAP RTP persisted despite the depletion of their RCK subunit. Thus, by interfering with reverse transcription, MOV10 inhibits IAP RTP, and this inhibition is independent of P bodies.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Virus de la Leucemia Murina de Moloney/metabolismo , Retroelementos , Transcripción Genética , Células HEK293 , Humanos , Inmunoprecipitación , Fenilalanina/química , ARN/metabolismo , ARN Interferente Pequeño/metabolismo , ARN de Transferencia/química , Secuencias Repetidas Terminales , Transfección , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA