Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 1544, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35318328

RESUMEN

Rhabdoid tumors (RT) are rare and highly aggressive pediatric neoplasms. Their epigenetically-driven intertumoral heterogeneity is well described; however, the cellular origin of RT remains an enigma. Here, we establish and characterize different genetically engineered mouse models driven under the control of distinct promoters and being active in early progenitor cell types with diverse embryonic onsets. From all models only Sox2-positive progenitor cells give rise to murine RT. Using single-cell analyses, we identify distinct cells of origin for the SHH and MYC subgroups of RT, rooting in early stages of embryogenesis. Intra- and extracranial MYC tumors harbor common genetic programs and potentially originate from fetal primordial germ cells (PGCs). Using PGC specific Smarcb1 knockout mouse models we validate that MYC RT originate from these progenitor cells. We uncover an epigenetic imbalance in MYC tumors compared to PGCs being sustained by epigenetically-driven subpopulations. Importantly, treatments with the DNA demethylating agent decitabine successfully impair tumor growth in vitro and in vivo. In summary, our work sheds light on the origin of RT and supports the clinical relevance of DNA methyltransferase inhibitors against this disease.


Asunto(s)
Tumor Rabdoide , Animales , Células Germinativas/patología , Humanos , Ratones , Tumor Rabdoide/genética , Tumor Rabdoide/patología , Proteína SMARCB1/genética , Análisis de la Célula Individual , Transcriptoma
2.
Int J Mol Sci ; 23(2)2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-35054996

RESUMEN

Inhibition of the dual function cell cycle and transcription kinase CDK7 is known to affect the viability of cancer cells, but the mechanisms underlying cell line-specific growth control remain poorly understood. Here, we employed a previously developed, highly specific small molecule inhibitor that non-covalently blocks ATP binding to CDK7 (LDC4297) to study the mechanisms underlying cell line-specific growth using a panel of genetically heterogeneous human pancreatic tumor lines as model system. Although LDC4297 diminished both transcription rates and CDK T-loop phosphorylation in a comparable manner, some PDAC lines displayed significantly higher sensitivity than others. We focused our analyses on two well-responsive lines (Mia-Paca2 and Panc89) that, however, showed significant differences in their viability upon extended exposure to limiting LDC4297 concentrations. Biochemical and RNAseq analysis revealed striking differences in gene expression and cell cycle control. Especially the downregulation of a group of cell cycle control genes, among them CDK1/2 and CDC25A/C, correlated well to the observed viability differences in Panc89 versus Mia-Paca2 cells. A parallel downregulation of regulatory pathways supported the hypothesis of a feedforward programmatic effect of CDK7 inhibitors, eventually causing hypersensitivity of PDAC lines.


Asunto(s)
Ciclo Celular/genética , Quinasas Ciclina-Dependientes/genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/metabolismo , Relación Dosis-Respuesta a Droga , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , FN-kappa B/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Quinasa Activadora de Quinasas Ciclina-Dependientes
3.
Nat Commun ; 11(1): 3409, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32641778

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is associated with high mortality and therapy resistance. Here, we show that low expression of κB-Ras GTPases is frequently detected in PDAC and correlates with higher histologic grade. In a model of KRasG12D-driven PDAC, loss of κB-Ras accelerates tumour development and shortens median survival. κB-Ras deficiency promotes acinar-to-ductal metaplasia (ADM) during tumour initiation as well as tumour progression through intrinsic effects on proliferation and invasion. κB-Ras proteins are also required for acinar regeneration after pancreatitis, demonstrating a general role in control of plasticity. Molecularly, upregulation of Ral GTPase activity and Sox9 expression underlies the observed phenotypes, identifying a previously unrecognized function of Ral signalling in ADM. Our results provide evidence for a tumour suppressive role of κB-Ras proteins and highlight low κB-Ras levels and consequent loss of Ral control as risk factors, thus emphasizing the necessity for therapeutic options that allow interference with Ral-driven signalling.


Asunto(s)
Células Acinares/metabolismo , Carcinoma Ductal Pancreático/genética , GTP Fosfohidrolasas/genética , Neoplasias Pancreáticas/genética , Pancreatitis/genética , Proteínas/genética , Células Acinares/patología , Anciano , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Femenino , GTP Fosfohidrolasas/metabolismo , Regulación de la Expresión Génica , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Estimación de Kaplan-Meier , Masculino , Metaplasia/genética , Metaplasia/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pancreatitis/metabolismo , Proteínas/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Proteínas de Unión al GTP ral/genética , Proteínas de Unión al GTP ral/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
4.
Nucleic Acids Res ; 47(6): 2793-2806, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30649478

RESUMEN

The TATA-box Binding Protein (TBP) plays a central role in regulating gene expression and is the first step in the process of pre-initiation complex (PIC) formation on promoter DNA. The lifetime of TBP at the promoter site is controlled by several cofactors including the Modifier of transcription 1 (Mot1), an essential TBP-associated ATPase. Based on ensemble measurements, Mot1 can use adenosine triphosphate (ATP) hydrolysis to displace TBP from DNA and various models for how this activity is coupled to transcriptional regulation have been proposed. However, the underlying molecular mechanism of Mot1 action is not well understood. In this work, the interaction of Mot1 with the DNA/TBP complex was investigated by single-pair Förster resonance energy transfer (spFRET). Upon Mot1 binding to the DNA/TBP complex, a transition in the DNA/TBP conformation was observed. Hydrolysis of ATP by Mot1 led to a conformational change but was not sufficient to efficiently disrupt the complex. SpFRET measurements of dual-labeled DNA suggest that Mot1's ATPase activity primes incorrectly oriented TBP for dissociation from DNA and additional Mot1 in solution is necessary for TBP unbinding. These findings provide a framework for understanding how the efficiency of Mot1's catalytic activity is tuned to establish a dynamic pool of TBP without interfering with stable and functional TBP-containing complexes.


Asunto(s)
Adenosina Trifosfatasas/fisiología , ADN de Hongos/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Factores Asociados con la Proteína de Unión a TATA/fisiología , Proteína de Unión a TATA-Box/química , Proteína de Unión a TATA-Box/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Catálisis , ADN de Hongos/química , Escherichia coli , Regulación Fúngica de la Expresión Génica , Modelos Moleculares , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Unión Proteica , Conformación Proteica , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Factores Asociados con la Proteína de Unión a TATA/química , Factores Asociados con la Proteína de Unión a TATA/metabolismo
5.
Biophys J ; 115(12): 2310-2326, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30527334

RESUMEN

Single-pair Förster resonance energy transfer (spFRET) has become an important tool for investigating conformational dynamics in biological systems. To extract dynamic information from the spFRET traces measured with total internal reflection fluorescence microscopy, we extended the hidden Markov model (HMM) approach. In our extended HMM analysis, we incorporated the photon-shot noise from camera-based systems into the HMM. Thus, the variance in Förster resonance energy transfer (FRET) efficiency of the various states, which is typically a fitted parameter, is explicitly included in the analysis estimated from the number of detected photons. It is also possible to include an additional broadening of the FRET state, which would then only reflect the inherent flexibility of the dynamic biological systems. This approach is useful when comparing the dynamics of individual molecules for which the total intensities vary significantly. We used spFRET with the extended HMM analysis to investigate the dynamics of TATA-box-binding protein (TBP) on promoter DNA in the presence of negative cofactor 2 (NC2). We compared the dynamics of two promoters as well as DNAs of different length and labeling location. For the adenovirus major late promoter, four FRET states were observed; three states correspond to different conformations of the DNA in the TBP-DNA-NC2 complex and a four-state model in which the complex has shifted along the DNA. The HMM analysis revealed that the states are connected via a linear, four-well model. For the H2B promoter, more complex dynamics were observed. By clustering the FRET states detected with the HMM analysis, we could compare the general dynamics observed for the two promoter sequences. We observed that the dynamics from a stretched DNA conformation to a bent conformation for the two promoters were similar, whereas the bent conformation of the TBP-DNA-NC2 complex for the H2B promoter is approximately three times more stable than for the adenovirus major late promoter.


Asunto(s)
ADN/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Cadenas de Markov , Proteína de Unión a TATA-Box/metabolismo , Factores de Transcripción/metabolismo , ADN/química , Modelos Moleculares , Conformación de Ácido Nucleico , Conformación Proteica , Proteína de Unión a TATA-Box/química , Factores de Transcripción/química
6.
Neurogenetics ; 19(4): 215-225, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30039206

RESUMEN

Charcot-Marie-Tooth disease (CMT) represents a heterogeneous group of hereditary peripheral neuropathies. We previously reported a CMT locus on chromosome 19q13.3 segregating with the disease in a large Costa Rican family with axonal neuropathy and autosomal recessive pattern of inheritance (CMT2B2). We proposed a homozygous missense variant in the Mediator complex 25 (MED25) gene as causative of the disease. Nevertheless, the fact that no other CMT individuals with MED25 variants were reported to date led us to reevaluate the original family. Using exome sequencing, we now identified a homozygous nonsense variant (p.Gln517ter) in the last exon of an adjacent gene, the polynucleotide kinase 3'-phosphatase (PNKP) gene. It encodes a DNA repair protein recently associated with recessive ataxia with oculomotor apraxia type 4 (AOA4) and microcephaly, seizures, and developmental delay (MCSZ). Subsequently, five unrelated Costa Rican CMT2 subjects initially identified as being heterozygous for the same MED25 variant were found to be also compound heterozygote for PNKP. All were heterozygous for the same variant found homozygous in the large family and a second one previously associated with ataxia (p.Thr408del). Detailed clinical reassessment of the initial family and the new individuals revealed in all an adult-onset slowly progressive CMT2 associated with signs of cerebellar dysfunction such as slurred speech and oculomotor involvement, but neither microcephaly, seizures, nor developmental delay. We propose that PKNP variants are the major causative variant for the CMT2 phenotype in these individuals and that the milder clinical manifestation is due to an allelic effect.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/genética , Enzimas Reparadoras del ADN/genética , Complejo Mediador/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Adulto , Sustitución de Aminoácidos/genética , Estudios de Casos y Controles , Consanguinidad , Costa Rica , Análisis Mutacional de ADN , Enzimas Reparadoras del ADN/química , Familia , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Mutación Missense , Linaje , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Polimorfismo de Nucleótido Simple
7.
Oncotarget ; 8(49): 84986-84995, 2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29156698

RESUMEN

Rhabdoid tumors are caused by the deletion of SMARCB1, whose protein encodes the SMARCB1 subunit of the chromatin remodeling complex SWI/SNF that is involved in global chromatin organization and gene expression control. Simultaneously inhibiting the main players involved in the deregulated transcription machinery is a promising option for preventing exaggerated tumor cell proliferation and survival as it may bypass compensatory mechanisms. In support of this hypothesis, we report efficient impairment of cellular proliferation and strong induction of cell death elicited by inhibition of bromodomain protein BRD4 and transcription kinase CDK9 using small molecular compounds. Combination of both compounds efficiently represses antiapoptotic genes and the oncogene MYC. Our results provide a novel approach for the treatment of RT.

8.
PLoS One ; 11(1): e0146648, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26745862

RESUMEN

CDK9 is the catalytic subunit of positive elongation factor b (P-TEFb) that controls the transition of RNA polymerase II (RNAPII) into elongation. CDK9 inhibitors block mRNA synthesis and trigger activation of the stress-sensitive p53 protein. This in turn induces transcription of CDKN1A (p21) and other cell cycle control genes. It is presently unclear if and how p53 circumvents a general P-TEFb-requirement when it activates its target genes. Our investigations using a panel of specific inhibitors reason for a critical role of CDK9 also in the case of direct inhibition of the kinase. At the prototypic p21 gene, the activator p53 initially accumulates at the pre-bound upstream enhancer followed-with significant delay-by de novo binding to a secondary enhancer site within the first intron of p21. This is accompanied by recruitment of the RNAPII initiation machinery to both elements. ChIP and functional analyses reason for a prominent role of CDK9 itself and elongation factor complexes PAF1c and SEC involved in pause and elongation control. It appears that the strong activation potential of p53 facilitates gene activation in the situation of global repression of RNAPII transcription. The data further underline the fundamental importance of CDK9 for class II gene transcription.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Polimerasa II/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Alineación de Secuencia , Factores de Transcripción , Activación Transcripcional , Proteína p53 Supresora de Tumor/genética
9.
Mol Cell Biol ; 34(19): 3675-88, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25047832

RESUMEN

Cyclin-dependent kinase 7 (CDK7) activates cell cycle CDKs and is a member of the general transcription factor TFIIH. Although there is substantial evidence for an active role of CDK7 in mRNA synthesis and associated processes, the degree of its influence on global and gene-specific transcription in mammalian species is unclear. In the current study, we utilize two novel inhibitors with high specificity for CDK7 to demonstrate a restricted but robust impact of CDK7 on gene transcription in vivo and in in vitro-reconstituted reactions. We distinguish between relative low- and high-dose responses and relate them to distinct molecular mechanisms and altered physiological responses. Low inhibitor doses cause rapid clearance of paused RNA polymerase II (RNAPII) molecules and sufficed to cause genome-wide alterations in gene expression, delays in cell cycle progression at both the G1/S and G2/M checkpoints, and diminished survival of human tumor cells. Higher doses and prolonged inhibition led to strong reductions in RNAPII carboxyl-terminal domain (CTD) phosphorylation, eventual activation of the p53 program, and increased cell death. Together, our data reason for a quantitative contribution of CDK7 to mRNA synthesis, which is critical for cellular homeostasis.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , Triazinas/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Neoplasias/genética , Fosforilación , Roscovitina , Quinasa Activadora de Quinasas Ciclina-Dependientes
10.
Int J Cancer ; 135(4): 989-95, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24420698

RESUMEN

Rhabdoid tumors are highly aggressive tumors occurring in infants and very young children. Despite multimodal and intensive therapy prognosis remains poor. Molecular analyses have uncovered several deregulated pathways, among them the CDK4/6-Rb-, the WNT- and the Sonic hedgehog (SHH) pathways. The SHH pathway is activated in rhabdoid tumors by GLI1 overexpression. Here, we demonstrate that arsenic trioxide (ATO) inhibits tumor cell growth of malignant rhabdoid tumors in vitro and in a mouse xenograft model by suppressing Gli1. Our data uncover ATO as a promising therapeutic approach to improve prognosis for rhabdoid tumor patients.


Asunto(s)
Antineoplásicos/farmacología , Arsenicales/farmacología , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Óxidos/farmacología , Tumor Rabdoide/tratamiento farmacológico , Factores de Transcripción/metabolismo , Animales , Apoptosis , Trióxido de Arsénico , Ciclo Celular , Proliferación Celular , Biología Computacional , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Pronóstico , Transducción de Señal , Proteína con Dedos de Zinc GLI1
11.
BMC Cancer ; 13: 286, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23764045

RESUMEN

BACKGROUND: Rhabdoid tumors are highly aggressive malignancies affecting infants and very young children. In many instances these tumors are resistant to conventional type chemotherapy necessitating alternative approaches. METHODS: Proliferation assays (MTT), apoptosis (propidium iodide/annexin V) and cell cycle analysis (DAPI), RNA expression microarrays and western blots were used to identify synergism of the HDAC (histone deacetylase) inhibitor SAHA with fenretinide, tamoxifen and doxorubicin in rhabdoidtumor cell lines. RESULTS: HDAC1 and HDAC2 are overexpressed in primary rhabdoid tumors and rhabdoid tumor cell lines. Targeting HDACs in rhabdoid tumors induces cell cycle arrest and apoptosis. On the other hand HDAC inhibition induces deregulated gene programs (MYCC-, RB program and the stem cell program) in rhabdoid tumors. These programs are in general associated with cell cycle progression. Targeting these activated pro-proliferative genes by combined approaches of HDAC-inhibitors plus fenretinide, which inhibits cyclinD1, exhibit strong synergistic effects on induction of apoptosis. Furthermore, HDAC inhibition sensitizes rhabdoid tumor cell lines to cell death induced by chemotherapy. CONCLUSION: Our data demonstrate that HDAC inhibitor treatment in combination with fenretinide or conventional chemotherapy is a promising tool for the treatment of chemoresistant rhabdoid tumors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores de Histona Desacetilasas/administración & dosificación , Histona Desacetilasas/metabolismo , Tumor Rabdoide/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Sinergismo Farmacológico , Fenretinida/administración & dosificación , Humanos , Ácidos Hidroxámicos/administración & dosificación , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Tumor Rabdoide/patología , Vorinostat
12.
Semin Cell Dev Biol ; 22(7): 735-40, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21864698

RESUMEN

The Mediator complex serves as an adaptor for regulatory factors, recruits and controls RNA polymerase II promotes preinitiation complex formation and functions post initiation. There is increasing evidence for further coordinating roles of the Mediator complex in chromatin. Here we summarize interactions with regulatory, general and accessory factors that function in transcription and chromatin.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Complejo Mediador/metabolismo , ARN Polimerasa II/metabolismo , Transcripción Genética , Animales , Humanos , ARN Polimerasa II/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Levaduras/genética , Levaduras/metabolismo
13.
Nat Struct Mol Biol ; 18(4): 404-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21378965

RESUMEN

Eukaryotic transcription is regulated by interactions between gene-specific activators and the coactivator complex Mediator. Here we report the NMR structure of the Mediator subunit Med25 (also called Arc92) activator interaction domain (ACID) and analyze the structural and functional interaction of ACID with the archetypical acidic transcription activator VP16. Unlike other known activator targets, ACID forms a seven-stranded ß-barrel framed by three helices. The VP16 subdomains H1 and H2 bind to opposite faces of ACID and cooperate during promoter-dependent activated transcription in a in vitro system. The activator-binding ACID faces are functionally required and conserved among higher eukaryotes. Comparison with published activator structures reveals that the VP16 activation domain uses distinct interaction modes to adapt to unrelated target surfaces and folds that evolved for activator binding.


Asunto(s)
Proteína Vmw65 de Virus del Herpes Simple/metabolismo , Complejo Mediador/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Complejo Mediador/química , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Activación Transcripcional
14.
Genome Biol ; 11(3): R33, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20230619

RESUMEN

BACKGROUND: The general transcription factor TFIIB and its antagonist negative cofactor 2 (NC2) are hallmarks of RNA polymerase II (RNAPII) transcription. Both factors bind TATA box-binding protein (TBP) at promoters in a mutually exclusive manner. Dissociation of NC2 is thought to be followed by TFIIB association and subsequent preinitiation complex formation. TFIIB dissociates upon RNAPII promoter clearance, thereby providing a specific measure for steady-state preinitiation complex levels. As yet, genome-scale promoter mapping of human TFIIB has not been reported. It thus remains elusive how human core promoters contribute to preinitiation complex formation in vivo. RESULTS: We compare target genes of TFIIB and NC2 in human B cells and analyze associated core promoter architectures. TFIIB occupancy is positively correlated with gene expression, with the vast majority of promoters being GC-rich and lacking defined core promoter elements. TATA elements, but not the previously in vitro defined TFIIB recognition elements, are enriched in some 4 to 5% of the genes. NC2 binds to a highly related target gene set. Nonetheless, subpopulations show strong variations in factor ratios: whereas high TFIIB/NC2 ratios select for promoters with focused start sites and conserved core elements, high NC2/TFIIB ratios correlate to multiple start-site promoters lacking defined core elements. CONCLUSIONS: TFIIB and NC2 are global players that occupy active genes. Preinitiation complex formation is independent of core elements at the majority of genes. TATA and TATA-like elements dictate TFIIB occupancy at a subset of genes. Biochemical data support a model in which preinitiation complex but not TBP-NC2 complex formation is regulated.


Asunto(s)
Complejos Multiproteicos/metabolismo , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , Proteína de Unión a TATA-Box/metabolismo , Factor de Transcripción TFIIB/metabolismo , Factores de Transcripción/metabolismo , Linfocitos B , Células Cultivadas , Inmunoprecipitación de Cromatina , Biología Computacional , Cartilla de ADN/genética , Humanos , Modelos Genéticos , Complejos Multiproteicos/genética , Fosfoproteínas/genética , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/metabolismo , Proteína de Unión a TATA-Box/genética , Factor de Transcripción TFIIB/genética , Factores de Transcripción/genética
15.
J Gen Virol ; 91(Pt 5): 1138-49, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20089804

RESUMEN

In cells infected with Kaposi's sarcoma-associated herpesvirus (KSHV), the activation of mitogen-activated protein kinase (MAPK) pathways plays a crucial role early after virus infection as well as during reactivation. In order to systematically identify viral proteins activating MAPK pathways in KSHV-infected cells, a clone collection of KSHV open reading frames (ORFs) was screened for induction of the serum response element (SRE), as SRE is induced by MAPKs. The strongest induction of the SRE was found with ORF73 (latency-associated nuclear antigen 1, or Lana-1), although weaker activation was also found with the kaposin B isoform, ORF54 (dUTPase) and ORF74 (G-protein-coupled receptor). The bipartite SRE is bound by a ternary complex consisting of serum response factor (SRF) and ternary complex factor. Lana-1 bound directly to SRF, but also to the MED25 (ARC92/ACID-1), MED15 (PCQAP) and MED23 (Sur-2) subunits of the Mediator complex, a multi-subunit transcriptional co-activator complex for RNA polymerase II. Lana-1-induced SRE activation was inhibited by the dominant-negative N-terminal domain of the MED25 mediator subunit, suggesting that this subunit mediates Lana-1-induced SRE activation. In summary, these data suggest a model in which Lana-1 acts as an adaptor between the transcription factor SRF and the basal transcriptional machinery.


Asunto(s)
Antígenos Virales/fisiología , Herpesvirus Humano 8/fisiología , Complejo Mediador/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Nucleares/fisiología , Elemento de Respuesta al Suero , Línea Celular , Genes Reporteros , Humanos , Luciferasas/metabolismo , Modelos Biológicos , Unión Proteica , Activación Transcripcional
16.
J Biol Chem ; 285(1): 188-96, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19901026

RESUMEN

The largest subunit of RNA polymerase II (RNAPII) C-terminal heptarepeat domain (CTD) is subject to phosphorylation during initiation and elongation of transcription by RNA polymerase II. Here we study the molecular mechanisms leading to phosphorylation of Ser-7 in the human enzyme. Ser-7 becomes phosphorylated before initiation of transcription at promoter regions. We identify cyclin-dependent kinase 7 (CDK7) as one responsible kinase. Phosphorylation of both Ser-5 and Ser-7 is fully dependent on the cofactor complex Mediator. A subform of Mediator associated with an active RNAPII is critical for preinitiation complex formation and CTD phosphorylation. The Mediator-RNAPII complex independently recruits TFIIB and CDK7 to core promoter regions. CDK7 phosphorylates Ser-7 selectively in the context of an intact preinitiation complex. CDK7 is not the only kinase that can modify Ser-7 of the CTD. ChIP experiments with chemical inhibitors provide evidence that other yet to be identified kinases further phosphorylate Ser-7 in coding regions.


Asunto(s)
Complejo Mediador/metabolismo , ARN Polimerasa II/química , ARN Polimerasa II/metabolismo , Secuencias Repetitivas de Aminoácido , Serina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , ADN/metabolismo , Células HeLa , Humanos , Células Jurkat , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Purinas/farmacología , Roscovitina , Moldes Genéticos , Factor de Transcripción TFIIH/metabolismo , Transcripción Genética/efectos de los fármacos , Quinasa Activadora de Quinasas Ciclina-Dependientes
17.
Neurogenetics ; 10(4): 275-87, 2009 10.
Artículo en Inglés | MEDLINE | ID: mdl-19290556

RESUMEN

Charcot-Marie-Tooth (CMT) disease is a clinically and genetically heterogeneous disorder. All mendelian patterns of inheritance have been described. We identified a homozygous p.A335V mutation in the MED25 gene in an extended Costa Rican family with autosomal recessively inherited Charcot-Marie-Tooth neuropathy linked to the CMT2B2 locus in chromosome 19q13.3. MED25, also known as ARC92 and ACID1, is a subunit of the human activator-recruited cofactor (ARC), a family of large transcriptional coactivator complexes related to the yeast Mediator. MED25 was identified by virtue of functional association with the activator domains of multiple cellular and viral transcriptional activators. Its exact physiological function in transcriptional regulation remains obscure. The CMT2B2-associated missense amino acid substitution p.A335V is located in a proline-rich region with high affinity for SH3 domains of the Abelson type. The mutation causes a decrease in binding specificity leading to the recognition of a broader range of SH3 domain proteins. Furthermore, Med25 is coordinately expressed with Pmp22 gene dosage and expression in transgenic mice and rats. These results suggest a potential role of this protein in the molecular etiology of CMT2B2 and suggest a potential, more general role of MED25 in gene dosage sensitive peripheral neuropathy pathogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Sustitución de Aminoácidos , Proteínas de Ciclo Celular , Enfermedad de Charcot-Marie-Tooth/genética , Complejo Mediador , Proteínas de la Mielina , Proteínas Nucleares , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Costa Rica , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Femenino , Dosificación de Gen , Genotipo , Humanos , Masculino , Complejo Mediador/química , Complejo Mediador/genética , Complejo Mediador/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Linaje , Conformación Proteica , Ratas
18.
J Biol Chem ; 284(14): 9382-93, 2009 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-19204005

RESUMEN

The negative cofactor 2 (NC2) is a protein complex composed of two subunits, NC2alpha and NC2beta, and plays a key role in transcription regulation. Here we investigate whether each subunit contains a nuclear localization signal (NLS) that permits individual crossing of the nuclear membrane or whether nuclear import of NC2alpha and NC2beta depends on heterodimerization. Our results from in vitro binding studies and transfection experiments in cultured cells show that each subunit contains a classical NLS (cNLS) that is recognized by the importin alpha/beta heterodimer. Regardless of the individual cNLSs the two NC2 subunits are translocated as a preassembled complex as co-transfection experiments with wild-type and cNLS-deficient NC2 subunits demonstrate. Ran-dependent binding of the nuclear export receptor Crm1/exportin 1 confirmed the presence of a leucine-rich nuclear export signal (NES) in NC2beta. In contrast, NC2alpha does not exhibit a NES. Our results from interspecies heterokaryon assays suggest that heterodimerization with NC2alpha masks the NES in NC2beta, which prevents nuclear export of the NC2 complex. A mutation in either one of the two cNLSs decreases the extent of importin alpha/beta-mediated nuclear import of the NC2 complex. In addition, the NC2 complex can enter the nucleus via a second pathway, facilitated by importin 13. Because importin 13 binds exclusively to the NC2 complex but not to the individual subunits this alternative import pathway depends on sequence elements distributed among the two subunits.


Asunto(s)
Fosfoproteínas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Línea Celular , Ácidos Grasos Insaturados/farmacología , Regulación de la Expresión Génica , Humanos , Carioferinas/metabolismo , Ratones , Señales de Localización Nuclear , Fosfoproteínas/genética , Multimerización de Proteína , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/genética , Proteína Exportina 1
20.
J Cell Biol ; 183(5): 769-76, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047459

RESUMEN

The multifunctional nuclear protein positive cofactor 4 (PC4) is involved in various cellular processes including transcription, replication, and chromatin organization. Recently, PC4 has been identified as a suppressor of oxidative mutagenesis in Escherichia coli and Saccharomyces cerevisiae. To investigate a potential role of PC4 in mammalian DNA repair, we used a combination of live cell microscopy, microirradiation, and fluorescence recovery after photobleaching analysis. We found a clear accumulation of endogenous PC4 at DNA damage sites introduced by either chemical agents or laser microirradiation. Using fluorescent fusion proteins and specific mutants, we demonstrated that the rapid recruitment of PC4 to laser-induced DNA damage sites is independent of poly(ADP-ribosyl)ation and gammaH2AX but depends on its single strand binding capacity. Furthermore, PC4 showed a high turnover at DNA damages sites compared with the repair factors replication protein A and proliferating cell nuclear antigen. We propose that PC4 plays a role in the early response to DNA damage by recognizing single-stranded DNA and may thus initiate or facilitate the subsequent steps of DNA repair.


Asunto(s)
Núcleo Celular/enzimología , Daño del ADN , Reparación del ADN , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Polimerasa II/metabolismo , Proteína de Replicación A/metabolismo , Factores de Transcripción/metabolismo , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/efectos de la radiación , Proteínas de Unión al ADN/genética , Recuperación de Fluorescencia tras Fotoblanqueo , Células HeLa , Histonas/metabolismo , Humanos , Cinética , Ratones , Ratones Noqueados , Microscopía Fluorescente , Mutación , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...