Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(4): 1687-1706, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36727434

RESUMEN

Positive transcription elongation factor b (P-TEFb) is the crucial player in RNA polymerase II (Pol II) pause release that has emerged as a promising target in cancer. Because single-agent therapy may fail to deliver durable clinical response, targeting of P-TEFb shall benefit when deployed as a combination therapy. We screened a comprehensive oncology library and identified clinically relevant antimetabolites and Mouse double minute 2 homolog (MDM2) inhibitors as top compounds eliciting p53-dependent death of colorectal cancer cells in synergy with selective inhibitors of P-TEFb. While the targeting of P-TEFb augments apoptosis by anti-metabolite 5-fluorouracil, it switches the fate of cancer cells by the non-genotoxic MDM2 inhibitor Nutlin-3a from cell-cycle arrest to apoptosis. Mechanistically, the fate switching is enabled by the induction of p53-dependent pro-apoptotic genes and repression of P-TEFb-dependent pro-survival genes of the PI3K-AKT signaling cascade, which stimulates caspase 9 and intrinsic apoptosis pathway in BAX/BAK-dependent manner. Finally, combination treatments trigger apoptosis of cancer cell spheroids. Together, co-targeting of P-TEFb and suppressors of intrinsic apoptosis could become a viable strategy to eliminate cancer cells.


Asunto(s)
Apoptosis , Factor B de Elongación Transcripcional Positiva , Proteínas Proto-Oncogénicas c-mdm2 , Proteína p53 Supresora de Tumor , Línea Celular Tumoral , Supervivencia Celular , Fosfatidilinositol 3-Quinasas/metabolismo , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteína p53 Supresora de Tumor/genética , Humanos
2.
Cell Commun Signal ; 19(1): 83, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34372855

RESUMEN

BACKGROUND: The positive transcription elongation factor b (P-TEFb) kinase activity is involved in the process of transcription. Cyclin-dependent kinase 9 (CDK9), a core component of P-TEFb, regulates the process of transcription elongation, which is associated with differentiation and apoptosis in many cancer types. Wogonin, a natural CDK9 inhibitor isolated from Scutellaria baicalensis. This study aimed to investigate the involved molecular mechanisms of wogonin on anti- chronic myeloid leukemia (CML) cells. MATERIALS AND METHODS: mRNA and protein levels were analysed by RT-qPCR and western blot. Flow cytometry was used to assess cell differentiation and apoptosis. Cell transfection, immunofluorescence analysis and co-immunoprecipitation (co-IP) assays were applied to address the potential regulatory mechanism of wogonin. KU-812 cells xenograft NOD/SCID mice model was used to assess and verify the mechanism in vivo. RESULTS: We reported that the anti-CML effects in K562, KU-812 and primary CML cells induced by wogonin were regulated by P-TEFb complex. We also confirmed the relationship between CDK9 and erythroid differentiation via knockdown the expression of CDK9. For further study the mechanism of erythroid differentiation induced by wogonin, co-IP experiments were used to demonstrate that wogonin increased the binding between GATA-1 and FOG-1 but decreased the binding between GATA-1 and RUNX1, which were depended on P-TEFb. Also, wogonin induced apoptosis and decreased the mRNA and protein levels of MCL-1 in KU-812 cells, which is the downstream of P-TEFb. In vivo studies showed wogonin had good anti-tumor effects in KU-812 xenografts NOD/ SCID mice model and decreased the proportion of human CD45+ cells in spleens of mice. We also verified that wogonin exhibited anti-CML effects through modulating P-TEFb activity in vivo. CONCLUSIONS: Our study indicated a special mechanism involving the regulation of P-TEFb kinase activity in CML cells, providing evidences for further application of wogonin in CML clinical treatment. Video Abstract.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/genética , Flavanonas/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Factor B de Elongación Transcripcional Positiva/genética , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Factor de Transcripción GATA1/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Terapia Molecular Dirigida , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/genética , Proteínas Nucleares/genética , Fosforilación/efectos de los fármacos , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Factores de Transcripción/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Molecules ; 25(4)2020 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-32075058

RESUMEN

The positive transcription elongation factor b (P-TEFb) was first identified as a general factor that stimulates transcription elongation by RNA polymerase II (RNAPII), but soon afterwards it turned out to be an essential cellular co-factor of human immunodeficiency virus (HIV) transcription mediated by viral Tat proteins. Studies on the mechanisms of Tat-dependent HIV transcription have led to radical advances in our knowledge regarding the mechanism of eukaryotic transcription, including the discoveries that P-TEFb-mediated elongation control of cellular transcription is a main regulatory step of gene expression in eukaryotes, and deregulation of P-TEFb activity plays critical roles in many human diseases and conditions in addition to HIV/AIDS. P-TEFb is now recognized as an attractive and promising therapeutic target for inflammation/autoimmune diseases, cardiac hypertrophy, cancer, infectious diseases, etc. In this review article, I will summarize our knowledge about basic P-TEFb functions, the regulatory mechanism of P-TEFb-dependent transcription, P-TEFb's involvement in biological processes and diseases, and current approaches to manipulating P-TEFb functions for the treatment of these diseases.


Asunto(s)
Infecciones por VIH/genética , Factor B de Elongación Transcripcional Positiva/genética , Transcripción Genética/efectos de los fármacos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Regulación Viral de la Expresión Génica/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , Humanos , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores
4.
Development ; 146(24)2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31784460

RESUMEN

Multipotent progenitor populations are necessary for generating diverse tissue types during embryogenesis. We show the RNA polymerase-associated factor 1 complex (Paf1C) is required to maintain multipotent progenitors of the neural crest (NC) lineage in zebrafish. Mutations affecting each Paf1C component result in near-identical NC phenotypes; alyron mutant embryos carrying a null mutation in paf1 were analyzed in detail. In the absence of zygotic paf1 function, definitive premigratory NC progenitors arise but fail to maintain expression of the sox10 specification gene. The mutant NC progenitors migrate aberrantly and fail to differentiate appropriately. Blood and germ cell progenitor development is affected similarly. Development of mutant NC could be rescued by additional loss of positive transcription elongation factor b (P-TEFb) activity, a key factor in promoting transcription elongation. Consistent with the interpretation that inhibiting/delaying expression of some genes is essential for maintaining progenitors, mutant embryos lacking the CDK9 kinase component of P-TEFb exhibit a surfeit of NC progenitors and their derivatives. We propose Paf1C and P-TEFb act antagonistically to regulate the timing of the expression of genes needed for NC development.


Asunto(s)
Linaje de la Célula/genética , Células Madre Multipotentes/fisiología , Cresta Neural/citología , Células-Madre Neurales/fisiología , Proteínas Nucleares/fisiología , Factor B de Elongación Transcripcional Positiva/fisiología , Factores de Transcripción/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Quinasa 9 Dependiente de la Ciclina/genética , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Células Madre Multipotentes/citología , Complejos Multiproteicos/genética , Complejos Multiproteicos/fisiología , Cresta Neural/fisiología , Células-Madre Neurales/citología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/metabolismo , ARN Polimerasa II/metabolismo , Factores de Transcripción/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
5.
Sci Adv ; 5(6): eaaw3593, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31183407

RESUMEN

Positive transcription elongation factor b (P-TEFb) functions as a central regulator of transcription elongation. Activation of P-TEFb occurs through its dissociation from the transcriptionally inactive P-TEFb/HEXIM1/7SK snRNP complex. However, the mechanisms of signal-regulated P-TEFb activation and its roles in human diseases remain largely unknown. Here, we demonstrate that cAMP-PKA signaling disrupts the inactive P-TEFb/HEXIM1/7SK snRNP complex by PKA-mediated phosphorylation of HEXIM1 at serine-158. The cAMP pathway plays central roles in the development of autosomal dominant polycystic kidney disease (ADPKD), and we show that P-TEFb is hyperactivated in mouse and human ADPKD kidneys. Genetic activation of P-TEFb promotes cyst formation in a zebrafish ADPKD model, while pharmacological inhibition of P-TEFb attenuates cyst development by suppressing the pathological gene expression program in ADPKD mice. Our study therefore elucidates a mechanism by which P-TEFb activation by cAMP-PKA signaling promotes cystogenesis in ADPKD.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Factor B de Elongación Transcripcional Positiva/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/química , Quistes/metabolismo , Quistes/patología , Modelos Animales de Enfermedad , Flavonoides/farmacología , Flavonoides/uso terapéutico , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Noqueados , Fosforilación , Piperidinas/farmacología , Piperidinas/uso terapéutico , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/metabolismo , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/genética , Unión Proteica , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/química , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Transducción de Señal , Canales Catiónicos TRPP/deficiencia , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Factores de Transcripción/metabolismo , Pez Cebra/metabolismo
6.
Cancer Discov ; 7(3): 302-321, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28108460

RESUMEN

Targeting the dysregulated BRAF-MEK-ERK pathway in cancer has increasingly emerged in clinical trial design. Despite clinical responses in specific cancers using inhibitors targeting BRAF and MEK, resistance develops often involving nongenomic adaptive bypass mechanisms. Inhibition of MEK1/2 by trametinib in patients with triple-negative breast cancer (TNBC) induced dramatic transcriptional responses, including upregulation of receptor tyrosine kinases (RTK) comparing tumor samples before and after one week of treatment. In preclinical models, MEK inhibition induced genome-wide enhancer formation involving the seeding of BRD4, MED1, H3K27 acetylation, and p300 that drives transcriptional adaptation. Inhibition of the P-TEFb-associated proteins BRD4 and CBP/p300 arrested enhancer seeding and RTK upregulation. BRD4 bromodomain inhibitors overcame trametinib resistance, producing sustained growth inhibition in cells, xenografts, and syngeneic mouse TNBC models. Pharmacologic targeting of P-TEFb members in conjunction with MEK inhibition by trametinib is an effective strategy to durably inhibit epigenomic remodeling required for adaptive resistance.Significance: Widespread transcriptional adaptation to pharmacologic MEK inhibition was observed in TNBC patient tumors. In preclinical models, MEK inhibition induces dramatic genome-wide modulation of chromatin, in the form of de novo enhancer formation and enhancer remodeling. Pharmacologic targeting of P-TEFb complex members at enhancers is an effective strategy to durably inhibit such adaptation. Cancer Discov; 7(3); 302-21. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 235.


Asunto(s)
Antineoplásicos/uso terapéutico , Elementos de Facilitación Genéticos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Piridonas/uso terapéutico , Pirimidinonas/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Azepinas/farmacología , Azepinas/uso terapéutico , Proteínas de Ciclo Celular , Línea Celular Tumoral , Metilación de ADN , Receptor con Dominio Discoidina 1/genética , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Epigénesis Genética , Femenino , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Ratones Endogámicos BALB C , Ratones SCID , Terapia Molecular Dirigida , Proteínas Nucleares/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Piridonas/farmacología , Pirimidinonas/farmacología , Interferencia de ARN , Factores de Transcripción/antagonistas & inhibidores , Triazoles/farmacología , Triazoles/uso terapéutico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Genesis ; 54(9): 470-82, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27488304

RESUMEN

Positive transcription elongation factor b (P-TEFb) is an RNA polymerase II kinase that phosphorylates Ser2 of the carboxyl-terminal domain and promotes the elongation phase of transcription. Despite the fact that P-TEFb has role in many cellular processes, the role of this kinase complex remains to be understood in early developmental events. In this study, using immunocytochemical analyses, we find that the P-TEFb components, Cyclin T1, CDK9, and its T-loop phosphorylated form, are localized to nuclear speckles, as well as in nucleoli in mouse germinal vesicle oocytes. Moreover, using fluorescence in situ hybridization, we show that in absence of CDK9 activity, nucleolar integration, as well as production of 28S rRNA is impaired in oocytes and embryos. We also present evidence indicating that P-TEFb kinase activity is essential for completion of mouse oocyte maturation and embryo development. Treatment with CDK9 inhibitor, flavopiridol resulted in metaphase I arrest in maturing oocytes. Inhibition of CDK9 kinase activity did not interfere with in vitro fertilization and pronuclear formation. However, when zygotes or 2-cell embryos were treated with flavopiridol only in their G2 phase of the cell cycle, development to the blastocyst stage was impaired. Inhibition of the CDK9 activity after embryonic genome activation resulted in failure to form normal blastocysts and aberrant phosphorylation of RNA polymerase II CTD. In all stages analyzed, treatment with flavopiridol abrogated global transcriptional activity. Collectively, our data suggest that P-TEFb kinase activity is crucial for oocyte maturation, embryo development, and regulation of global RNA transcription in mouse early development.


Asunto(s)
Blastocisto/metabolismo , Oogénesis , Factor B de Elongación Transcripcional Positiva/metabolismo , Transcriptoma , Animales , Blastocisto/efectos de los fármacos , Células Cultivadas , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/metabolismo , Femenino , Flavonoides/farmacología , Fase G2 , Ratones , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Piperidinas/farmacología , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , ARN Ribosómico 28S/metabolismo
8.
Science ; 352(6289): aad7993, 2016 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-27127234

RESUMEN

The host innate immune response is the first line of defense against pathogens and is orchestrated by the concerted expression of genes induced by microbial stimuli. Deregulated expression of these genes is linked to the initiation and progression of diseases associated with exacerbated inflammation. We identified topoisomerase 1 (Top1) as a positive regulator of RNA polymerase II transcriptional activity at pathogen-induced genes. Depletion or chemical inhibition of Top1 suppresses the host response against influenza and Ebola viruses as well as bacterial products. Therapeutic pharmacological inhibition of Top1 protected mice from death in experimental models of lethal inflammation. Our results indicate that Top1 inhibition could be used as therapy against life-threatening infections characterized by an acutely exacerbated immune response.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Inflamación/tratamiento farmacológico , Inflamación/genética , Inhibidores de Topoisomerasa I/uso terapéutico , Transcripción Genética/efectos de los fármacos , Animales , Azepinas/farmacología , Azepinas/uso terapéutico , Camptotecina/farmacología , Camptotecina/uso terapéutico , Ebolavirus , Flavonoides/farmacología , Flavonoides/uso terapéutico , Células HEK293 , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Inmunidad Innata , Inflamación/microbiología , Virus de la Influenza A , Interferón beta/inmunología , Ratones , Ratones Endogámicos C57BL , Piperidinas/farmacología , Piperidinas/uso terapéutico , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , ARN Polimerasa II/metabolismo , Virus Sendai , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus , Inhibidores de Topoisomerasa I/farmacología , Topotecan/uso terapéutico , Triazoles/farmacología , Triazoles/uso terapéutico
9.
PLoS One ; 11(3): e0152254, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27011207

RESUMEN

Positive transcription elongation factor b (P-TEFb) is a RNA polymerase II carboxyl-terminal domain (Pol II CTD) kinase that phosphorylates Ser2 of the CTD and promotes the elongation phase of transcription. Despite the fact that P-TEFb has role in many cellular processes, the role of this kinase complex remains to be understood in mammalian early developmental events. In this study, using immunocytochemical analyses, we found that the P-TEFb components, CDK9, Cyclin T1 and Cyclin T2 were localized to nuclear speckles, as well as in nucleolar-like bodies in pig germinal vesicle oocytes. Using nascent RNA labeling and small molecule inhibitors, we showed that inhibition of CDK9 activity abolished the transcription of GV oocytes globally. Moreover, using fluorescence in situ hybridization, in absence of CDK9 kinase activity the production of ribosomal RNAs was impaired. We also presented the evidences indicating that P-TEFb kinase activity is essential for resumption of oocyte meiosis and embryo development. Treatment with CDK9 inhibitors resulted in germinal vesicle arrest in maturing oocytes in vitro. Inhibition of CDK9 kinase activity did not interfere with in vitro fertilization and pronuclear formation. However, when in vitro produced zygotes were treated with CDK9 inhibitors, their development beyond the 4-cell stage was impaired. In these embryos, inhibition of CDK9 abrogated global transcriptional activity and rRNA production. Collectively, our data suggested that P-TEFb kinase activity is crucial for oocyte maturation, embryo development and regulation of RNA transcription in pig.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/genética , Oocitos/metabolismo , Factor B de Elongación Transcripcional Positiva/genética , Transcripción Genética , Animales , Ciclina T/genética , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/biosíntesis , Embrión de Mamíferos , Desarrollo Embrionario , Femenino , Fertilización In Vitro , Genoma , Técnicas de Maduración In Vitro de los Oocitos , Meiosis/genética , Oocitos/crecimiento & desarrollo , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/biosíntesis , Porcinos
10.
Nucleic Acids Res ; 43(17): 8258-67, 2015 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-26202968

RESUMEN

Phosphorylation of the C-terminal domain of the largest subunit of RNA polymerase II (Pol II), especially Ser2 and Ser5 residues, plays important roles in transcription and mRNA processing, including 5' end capping, splicing and 3' end processing. These phosphorylation events stimulate mRNA processing, however, it is not clear whether splicing activity affects the phosphorylation status of Pol II. In this study, we found that splicing inhibition by potent splicing inhibitors spliceostatin A (SSA) and pladienolide B or by antisense oligos against snRNAs decreased phospho-Ser2 level, but had little or no effects on phospho-Ser5 level. In contrast, transcription and translation inhibitors did not decrease phospho-Ser2 level, therefore inhibition of not all the gene expression processes cause the decrease of phospho-Ser2. SSA treatment caused early dissociation of Pol II and decrease in phospho-Ser2 level of chromatin-bound Pol II, suggesting that splicing inhibition causes downregulation of phospho-Ser2 through at least these two mechanisms.


Asunto(s)
ARN Polimerasa II/metabolismo , Empalme del ARN , Serina/metabolismo , Cromatina/metabolismo , Regulación hacia Abajo , Células HeLa , Humanos , Oligonucleótidos Antisentido , Fosforilación , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Estructura Terciaria de Proteína , Piranos/farmacología , ARN Polimerasa II/química , Empalme del ARN/efectos de los fármacos , ARN Nuclear Pequeño/antagonistas & inhibidores , Compuestos de Espiro/farmacología
11.
Cell Mol Life Sci ; 72(19): 3599-620, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26043973

RESUMEN

Germ cells must transmit genetic information across generations, and produce gametes while also maintaining the potential to form all cell types after fertilization. Preventing the activation of somatic programs is, therefore, crucial to the maintenance of germ cell identity. Studies in Caenorhabditis elegans, Drosophila melanogaster, and mouse have revealed both similarities and differences in how somatic gene expression is repressed in germ cells, thereby preventing their conversion into somatic tissues. This review will focus on recent developments in our understanding of how global or gene-specific transcriptional repression, chromatin regulation, and translational repression operate in the germline to maintain germ cell identity and repress somatic differentiation programs.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Cromatina/fisiología , Metilación de ADN , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Germinativas/citología , Modelos Biológicos , Animales , Caenorhabditis elegans , Drosophila , Proteínas de Drosophila/metabolismo , Humanos , Ratones , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , ARN Polimerasa II/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Factores de Transcripción SOXF/metabolismo , Especificidad de la Especie
12.
Curr Top Microbiol Immunol ; 389: 121-45, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25731772

RESUMEN

Antiretroviral therapy (ART) potently suppresses HIV-1 replication, but the virus persists in quiescent infected CD4(+)T cells as a latent integrated provirus, and patients must indefinitely remain on therapy. If ART is terminated, these integrated proviruses can reactivate, driving new rounds of infection. A functional cure for HIV requires eliminating low-level ongoing viral replication that persists in certain tissue sanctuaries and preventing viral reactivation. The HIV Tat protein plays an essential role in HIV transcription by recruiting the kinase activity of the P-TEFb complex to the viral mRNA's stem-bulge-loop structure, TAR, activating transcriptional elongation. Because the Tat-mediated transactivation cascade is critical for robust HIV replication, the Tat/TAR/P-TEFb complex is one of the most attractive targets for drug development. Importantly, compounds that interfere with transcription could impair viral reactivation, low-level ongoing replication, and replenishment of the latent reservoir, thereby reducing the size of the latent reservoir pool. Here, we discuss the potential importance of transcriptional inhibitors in the treatment of latent HIV-1 disease and review recent findings on targeting Tat, TAR, and P-TEFb individually or as part of a complex. Finally, we discuss the impact of extracellular Tat in HIV-associated neurocognitive disorders and cancers.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Fármacos Anti-VIH/farmacología , Duplicado del Terminal Largo de VIH/efectos de los fármacos , VIH-1 , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/antagonistas & inhibidores , Humanos
13.
Nat Rev Mol Cell Biol ; 16(3): 167-77, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25693130

RESUMEN

Recent advances in sequencing techniques that measure nascent transcripts and that reveal the positioning of RNA polymerase II (Pol II) have shown that the pausing of Pol II in promoter-proximal regions and its release to initiate a phase of productive elongation are key steps in transcription regulation. Moreover, after the release of Pol II from the promoter-proximal region, elongation rates are highly dynamic throughout the transcription of a gene, and vary on a gene-by-gene basis. Interestingly, Pol II elongation rates affect co-transcriptional processes such as splicing, termination and genome stability. Increasing numbers of factors and regulatory mechanisms have been associated with the steps of transcription elongation by Pol II, revealing that elongation is a highly complex process. Elongation is thus now recognized as a key phase in the regulation of transcription by Pol II.


Asunto(s)
Proteínas de Drosophila/genética , Genoma , Neoplasias/genética , Proteínas Nucleares/genética , Factor B de Elongación Transcripcional Positiva/genética , ARN Polimerasa II/genética , Elongación de la Transcripción Genética , Factores de Transcripción/genética , Animales , Antineoplásicos/uso terapéutico , Proteínas de Ciclo Celular , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Inestabilidad Genómica , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , ARN Polimerasa II/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo
14.
Anticancer Res ; 34(12): 6981-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25503124

RESUMEN

BACKGROUND: Positive transcription elongation factor-b (P-TEFb) is a complex containing CDK9 and a cyclin (T1, T2 or K). The effect of inhibition of P-TEFb by 5,6-dichloro-l-ß-D-ribofuranosyl benzimidazole (DRB) on cell radiosensitivity and the underlying mechanisms were investigated. MATERIALS AND METHODS: Six human cancer cell lines were subjected to (3)H-uridine incorporation, cell viability and clonogenic cell survival assays; cell-cycle redistribution and apoptosis assay; western blots and nuclear 53BP1 foci analysis after exposing the cells to DRB with/without γ-radiation. RESULTS: DRB suppressed colony formation and enhanced radiosensitivity of all cell lines. DRB caused a further increase in radiation-induced apoptosis and cell-cycle redistribution depending on p53 status. DRB prolonged the presence of radiation-induced nuclear p53 binding protein-1 (53BP1) foci and suppressed the expression of sirtuin-1 (SIRT1) and casein kinase 2-alpha (CK2α), suggesting an inhibition of DNA repair processes. CONCLUSION: Our findings indicate that DRB has the potential to increase the efficacy of radiotherapy and warrants further investigation using in vivo tumor models.


Asunto(s)
Quinasa de la Caseína II/antagonistas & inhibidores , Diclororribofuranosil Benzoimidazol/farmacología , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Sirtuina 1/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Quinasa de la Caseína II/biosíntesis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/efectos de la radiación , Sirtuina 1/biosíntesis , Proteína 1 de Unión al Supresor Tumoral P53
15.
PLoS One ; 9(9): e107654, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25233083

RESUMEN

The Herpes Simplex Virus 1 (HSV-1)-encoded ICP22 protein plays an important role in viral infection and affects expression of host cell genes. ICP22 is known to reduce the global level of serine (Ser)2 phosphorylation of the Tyr1Ser2Pro3Thr4Ser5Pro6Ser7 heptapeptide repeats comprising the carboxy-terminal domain (CTD) of the large subunit of RNA polymerase (pol) II. Accordingly, ICP22 is thought to associate with and inhibit the activity of the positive-transcription elongation factor b (P-TEFb) pol II CTD Ser2 kinase. We show here that ICP22 causes loss of CTD Ser2 phosphorylation from pol II engaged in transcription of protein-coding genes following ectopic expression in HeLa cells and that recombinant ICP22 interacts with the CDK9 subunit of recombinant P-TEFb. ICP22 also interacts with pol II in vitro. Residues 193 to 256 of ICP22 are sufficient for interaction with CDK9 and inhibition of pol II CTD Ser2 phosphorylation but do not interact with pol II. These results indicate that discrete regions of ICP22 interact with either CDK9 or pol II and that ICP22 interacts directly with CDK9 to inhibit expression of host cell genes.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Herpesvirus Humano 1/genética , Proteínas Inmediatas-Precoces/metabolismo , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , ARN Polimerasa II/antagonistas & inhibidores , Línea Celular Tumoral , Células HeLa , Humanos , Fosforilación , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Proteínas Quinasas/metabolismo , ARN Polimerasa II/genética , Transcripción Genética/genética
16.
Med Sci (Paris) ; 30(8-9): 797-802, 2014.
Artículo en Francés | MEDLINE | ID: mdl-25174758

RESUMEN

The transcription factor CTIP2 (BCL11B) is a multifunctional protein involved in numerous cell physiological processes. To date, many molecular mechanisms underlying this process have been discovered, which highlighted the importance of the epigenetic regulation of genes and the regulation of the elongation factor P-TEFb. Furthermore studies of the deregulation of CTIP2 showed the association of CTIP2 to numerous pathologies including cancer and cardiac hypertrophy. A better comprehension of the physiopathology of these diseases might lead to the design of therapeutical strategies intending to prevent CTIP2 deregulation. Moreover, CTIP2 and its associated proteins constitute potential targets in strategies aiming to reduce and/or purge HIV-1 cell reservoirs.


Asunto(s)
Terapia Molecular Dirigida , Proteínas Represoras/fisiología , Proteínas Supresoras de Tumor/fisiología , Síndrome de Inmunodeficiencia Adquirida/genética , Síndrome de Inmunodeficiencia Adquirida/terapia , Síndrome de Inmunodeficiencia Adquirida/virología , Animales , Cardiomegalia/genética , Cardiomegalia/terapia , Epigénesis Genética , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , VIH-1 , Humanos , Neoplasias/genética , Neoplasias/terapia , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Latencia del Virus/genética
17.
PLoS Genet ; 9(11): e1003897, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24244179

RESUMEN

Molecular mechanisms for the establishment of transcriptional memory are poorly understood. 5,6-dichloro-1-D-ribofuranosyl-benzimidazole (DRB) is a P-TEFb kinase inhibitor that artificially induces the poised RNA polymerase II (RNAPII), thereby manifesting intermediate steps for the establishment of transcriptional activation. Here, using genetics and DRB, we show that mammalian Absent, small, or homeotic discs 1-like (Ash1l), a member of the trithorax group proteins, methylates Lys36 of histone H3 to promote the establishment of Hox gene expression by counteracting Polycomb silencing. Importantly, we found that Ash1l-dependent Lys36 di-, tri-methylation of histone H3 in a coding region and exclusion of Polycomb group proteins occur independently of transcriptional elongation in embryonic stem (ES) cells, although both were previously thought to be consequences of transcription. Genome-wide analyses of histone H3 Lys36 methylation under DRB treatment have suggested that binding of the retinoic acid receptor (RAR) to a certain genomic region promotes trimethylation in the RAR-associated gene independent of its ongoing transcription. Moreover, DRB treatment unveils a parallel response between Lys36 methylation of histone H3 and occupancy of either Tip60 or Mof in a region-dependent manner. We also found that Brg1 is another key player involved in the response. Our results uncover a novel regulatory cascade orchestrated by Ash1l with RAR and provide insights into mechanisms underlying the establishment of the transcriptional activation that counteracts Polycomb silencing.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Proteínas del Grupo Polycomb/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Factores de Elongación Transcripcional/genética , Animales , Cromatina/genética , Proteínas de Unión al ADN/genética , Diclororribofuranosil Benzoimidazol/farmacología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Silenciador del Gen , Histona Acetiltransferasas/metabolismo , N-Metiltransferasa de Histona-Lisina , Histonas/genética , Humanos , Lisina/genética , Metilación , Proteínas Nucleares/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/metabolismo , ARN Polimerasa II/genética , Factores de Transcripción/genética , Factores de Elongación Transcripcional/metabolismo
18.
PLoS One ; 8(8): e72823, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977357

RESUMEN

Hexamethylene bisacetamide inducible protein 1 (HEXIM1) is best known as the inhibitor of positive transcription elongation factor b (P-TEFb), which is composed of cyclin-dependent kinase 9 (CDK9)/cyclin T1. P-TEFb is an essential regulator for the transcriptional elongation by RNA polymerase II. A genome-wide study using human embryonic stem cells shows that most mRNA synthesis is regulated at the stage of transcription elongation, suggesting a possible role for P-TEFb/HEXIM1 in the gene regulation of stem cells. In this report, we detected a marked increase in HEXIM1 protein levels in the differentiated human pluripotent stem cells (hPSCs) induced by LY294002 treatment. Since no changes in CDK9 and cyclin T1 were observed in the LY294002-treated cells, increased levels of HEXIM1 might lead to inhibition of P-TEFb activity. However, treatment with a potent P-TEFb inhibiting compound, flavopiridol, failed to induce hPSC differentiation, ruling out the possible requirement for P-TEFb kinase activity in hPSC differentiation. Conversely, differentiation was observed when hPSCs were incubated with hexamethylene bisacetamide, a HEXIM1 inducing reagent. The involvement of HEXIM1 in the regulation of hPSCs was further supported when overexpression of HEXIM1 concomitantly induced hPSC differentiation. Collectively, our study demonstrates a novel role of HEXIM1 in regulating hPSC fate through a P-TEFb-independent pathway.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas de Unión al ARN/metabolismo , Acetamidas/farmacología , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Ciclina T/metabolismo , Quinasa 9 Dependiente de la Ciclina/metabolismo , Ectodermo/citología , Flavonoides/farmacología , Humanos , Mesodermo/citología , Piperidinas/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Factor B de Elongación Transcripcional Positiva/metabolismo , Factores de Transcripción , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
19.
Nucleic Acids Res ; 40(13): 5988-6000, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22492511

RESUMEN

The MYB proto-oncogene is expressed in most estrogen receptor-positive (ERα(+)) breast tumors and cell lines. Expression of MYB is controlled, in breast cancer and other cell types, by a transcriptional pausing mechanism involving an attenuation site located ∼1.7 kb downstream from the transcription start site. In breast cancer cells, ligand-bound ERα binds close to, and drives transcription beyond this attenuation site, allowing synthesis of complete transcripts. However, little is known, in general, about the factors involved in relieving transcriptional attenuation, or specifically how ERα coordinates such factors to promote transcriptional elongation. Using cyclin dependent kinase 9 (CDK9) inhibitors, reporter gene assays and measurements of total and intronic MYB transcription, we show that functionally active CDK9 is required for estrogen-dependent transcriptional elongation. We further show by ChIP and co-immunoprecipitation studies that the P-TEFb complex (CDK9/CyclinT1) is recruited to the attenuation region by ligand-bound ERα, resulting in increased RNA polymerase II Ser-2 phosphorylation. These data provide new insights into MYB regulation, and given the critical roles of MYB in tumorigenesis, suggest targeting MYB elongation as potential therapeutic strategy.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Genes myb , Factor B de Elongación Transcripcional Positiva/metabolismo , Transcripción Genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Células HEK293 , Humanos , Intrones , Fosforilación , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Proto-Oncogenes Mas , ARN Polimerasa II/metabolismo , Elementos Reguladores de la Transcripción
20.
Eur J Med Chem ; 46(9): 4289-94, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21777997

RESUMEN

Small molecule inhibitors of cyclin-dependent kinases (CDK) have been developed as anticancer drugs with cytostatic and cytotoxic properties, but some of them have also been shown to limit angiogenesis. Here, we report that the 3,5-diaminopyrazole CAN508 inhibits endothelial cell migration and tube formation. In addition, it reduces phosphorylation of the C-terminus of RNA polymerase II and inhibits mRNA synthesis in endothelial cells, in accordance with previous observations that it has high selectivity towards the positive transcriptional regulator P-TEFb. Moreover, CAN508 reduces expression of vascular endothelial growth factor by several human cancer cell lines. The findings suggest that P-TEFb may be an attractive target for anti-angiogenic therapy.


Asunto(s)
Compuestos Azo/farmacología , Neovascularización Patológica/prevención & control , Factor B de Elongación Transcripcional Positiva/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Células Cultivadas , Humanos , Fosforilación , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA