Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 14(1): e0203577, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30703085

RESUMEN

RB-E2F transcriptional control plays a key role in regulating the timing of cell cycle progression from G1 to S-phase in response to growth factor stimulation. Despite this role, it is genetically dispensable for cell cycle exit in primary fibroblasts in response to growth arrest signals. Mice engineered to be defective for RB-E2F transcriptional control at cell cycle genes were also found to live a full lifespan with no susceptibility to cancer. Based on this background we sought to probe the vulnerabilities of RB-E2F transcriptional control defects found in Rb1R461E,K542E mutant mice (Rb1G) through genetic crosses with other mouse strains. We generated Rb1G/G mice in combination with Trp53 and Cdkn1a deficiencies, as well as in combination with KrasG12D. The Rb1G mutation enhanced Trp53 cancer susceptibility, but had no effect in combination with Cdkn1a deficiency or KrasG12D. Collectively, this study indicates that compromised RB-E2F transcriptional control is not uniformly cancer enabling, but rather has potent oncogenic effects when combined with specific vulnerabilities.


Asunto(s)
Factores de Transcripción E2F/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Proteína de Retinoblastoma/genética , Animales , Carcinogénesis/genética , Ciclo Celular/genética , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Fibroblastos , Predisposición Genética a la Enfermedad , Humanos , Antígeno Ki-67/análisis , Ratones , Ratones Transgénicos , Mutación , Neoplasias/patología , Cultivo Primario de Células , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteína de Retinoblastoma/metabolismo , Transducción de Señal/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Cell Div ; 12: 3, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28293272

RESUMEN

BACKGROUND: The G1-S phase transition is critical to maintaining proliferative control and preventing carcinogenesis. The retinoblastoma tumor suppressor is a key regulator of this step in the cell cycle. RESULTS: Here we use a structure-function approach to evaluate the contributions of multiple protein interaction surfaces on pRB towards cell cycle regulation. SAOS2 cell cycle arrest assays showed that disruption of three separate binding surfaces were necessary to inhibit pRB-mediated cell cycle control. Surprisingly, mutation of some interaction surfaces had no effect on their own. Rather, they only contributed to cell cycle arrest in the absence of other pRB dependent arrest functions. Specifically, our data shows that pRB-E2F interactions are competitive with pRB-CDH1 interactions, implying that interchangeable growth arrest functions underlie pRB's ability to block proliferation. Additionally, disruption of similar cell cycle control mechanisms in genetically modified mutant mice results in ectopic DNA synthesis in the liver. CONCLUSIONS: Our work demonstrates that pRB utilizes a network of mechanisms to prevent cell cycle entry. This has important implications for the use of new CDK4/6 inhibitors that aim to activate this proliferative control network.

3.
Mol Cell Biol ; 37(2)2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27821477

RESUMEN

The mammalian G1-S phase transition is controlled by the opposing forces of cyclin-dependent kinases (CDK) and the retinoblastoma protein (pRB). Here, we present evidence for systems-level control of cell cycle arrest by pRB-E2F and p27-CDK regulation. By introducing a point mutant allele of pRB that is defective for E2F repression (Rb1G) into a p27KIP1 null background (Cdkn1b-/-), both E2F transcriptional repression and CDK regulation are compromised. These double-mutant Rb1G/G; Cdkn1b-/- mice are viable and phenocopy Rb1+/- mice in developing pituitary adenocarcinomas, even though neither single mutant strain is cancer prone. Combined loss of pRB-E2F transcriptional regulation and p27KIP1 leads to defective proliferative control in response to various types of DNA damage. In addition, Rb1G/G; Cdkn1b-/- fibroblasts immortalize faster in culture and more frequently than either single mutant genotype. Importantly, the synthetic DNA damage arrest defect caused by Rb1G/G; Cdkn1b-/- mutations is evident in the developing intermediate pituitary lobe where tumors ultimately arise. Our work identifies a unique relationship between pRB-E2F and p27-CDK control and offers in vivo evidence that pRB is capable of cell cycle control through E2F-independent effects.


Asunto(s)
Puntos de Control del Ciclo Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Transcripción E2F/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteína de Retinoblastoma/metabolismo , Transcripción Genética , Animales , Línea Celular Transformada , Medio de Cultivo Libre de Suero , ADN/biosíntesis , Daño del ADN , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Ratones , Mutación/genética , Estrés Oxidativo , Hipófisis/embriología , Hipófisis/metabolismo , Biosíntesis de Proteínas/genética , Estabilidad Proteica , Tolerancia a Radiación
4.
J Mol Biol ; 428(20): 3960-3971, 2016 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-27567532

RESUMEN

The human pocket proteins retinoblastoma (Rb), p107, and p130 are critical negative regulators of the cell cycle and contribute to tumor suppression. While strong structural conservation within the pocket protein family provides for some functional redundancy, important differences have been observed and may underlie the reason that Rb is a uniquely potent tumor suppressor. It has been proposed that distinct pocket protein activities are mediated by their different E2F transcription factor binding partners. In humans, Rb binds E2F1-E2F5, whereas p107 and p130 almost exclusively associate with E2F4 and E2F5. To identify the molecular determinants of this specificity, we compared the crystal structures of Rb and p107 pocket domains and identified several key residues that contribute to E2F selectivity in the pocket family. Mutation of these residues in p107 to match the analogous residue in Rb results in an increase in affinity for E2F1 and E2F2 and an increase in the ability of p107 to inhibit E2F2 transactivation. Additionally, we investigated how phosphorylation by Cyclin-dependent kinase on distinct residues regulates p107 affinity for the E2F4 transactivation domain. We found that phosphorylation of residues S650 and S975 weakens the E2F4 transactivation domain binding. Our data reveal molecular features of pocket proteins that are responsible for their similarities and differences in function and regulation.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F4/metabolismo , Factor de Transcripción E2F5/metabolismo , Proteína de Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Cristalografía por Rayos X , Análisis Mutacional de ADN , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteína de Retinoblastoma/química , Proteína p107 Similar a la del Retinoblastoma/química , Proteína p107 Similar a la del Retinoblastoma/genética , Especificidad por Sustrato
5.
Methods Mol Biol ; 1342: 91-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26254919

RESUMEN

A fundamental need in the analysis of the cell cycle is the ability to isolate relatively homogeneous populations of cells in different phases. This is complicated by the variable proliferative properties and responses to synchronizing methods of different cancer-derived cell lines. Paradoxically, cell lines with genetic defects in cell cycle control are sometimes chosen because they are amenable to chemical synchronization. Embryonic fibroblasts from mice present the opportunity to study the effects of defined genetic modifications on a normal cell cycle. However, synchronization of these cells has often been challenging. In this chapter we outline three basic protocols for isolating mouse fibroblasts at the G1-to-S-phase transition, in S phase, and during mitosis.


Asunto(s)
Ciclo Celular , Separación Celular/métodos , Embrión de Mamíferos/citología , Fibroblastos/citología , Animales , Femenino , Fase G1 , Ratones , Mitosis , Embarazo , Fase S
6.
Methods Mol Biol ; 1170: 449-61, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24906329

RESUMEN

The G1/S-phase restriction point is an important landmark in the mammalian cell division cycle. The key regulator of the G1/S transition is the retinoblastoma gene product (pRB). It prevents the transcription of genes required for S-phase progression by repressing E2F transcription factors. An increase in Cdk phosphorylation of pRB causes the release of E2F transcription factors and advancement into S phase. Here we describe two simple techniques used to assess pRB phosphorylation and E2F transcription during G1/S progression.


Asunto(s)
Factores de Transcripción E2F/metabolismo , Fase G1 , Proteína de Retinoblastoma/metabolismo , Activación Transcripcional , Animales , Diseño de Equipo , Humanos , Immunoblotting/métodos , Hibridación de Ácido Nucleico/métodos , Fosforilación , ARN/genética , ARN/aislamiento & purificación
7.
Mol Cell Biol ; 34(12): 2221-34, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24710275

RESUMEN

Mammalian DREAM is a conserved protein complex that functions in cellular quiescence. DREAM contains an E2F, a retinoblastoma (RB)-family protein, and the MuvB core (LIN9, LIN37, LIN52, LIN54, and RBBP4). In mammals, MuvB can alternatively bind to BMYB to form a complex that promotes mitotic gene expression. Because BMYB-MuvB is essential for proliferation, loss-of-function approaches to study MuvB have generated limited insight into DREAM function. Here, we report a gene-targeted mouse model that is uniquely deficient for DREAM complex assembly. We have targeted p107 (Rbl1) to prevent MuvB binding and combined it with deficiency for p130 (Rbl2). Our data demonstrate that cells from these mice preferentially assemble BMYB-MuvB complexes and fail to repress transcription. DREAM-deficient mice show defects in endochondral bone formation and die shortly after birth. Micro-computed tomography and histology demonstrate that in the absence of DREAM, chondrocytes fail to arrest proliferation. Since DREAM requires DYRK1A (dual-specificity tyrosine phosphorylation-regulated protein kinase 1A) phosphorylation of LIN52 for assembly, we utilized an embryonic bone culture system and pharmacologic inhibition of (DYRK) kinase to demonstrate a similar defect in endochondral bone growth. This reveals that assembly of mammalian DREAM is required to induce cell cycle exit in chondrocytes.


Asunto(s)
Condrocitos/citología , Condrocitos/metabolismo , Mamíferos/metabolismo , Complejos Multiproteicos/metabolismo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Harmina/farmacología , Humanos , Ratones , Ratones Mutantes , Modelos Animales , Datos de Secuencia Molecular , Complejos Multiproteicos/química , Mutación/genética , Osteogénesis/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Tibia/efectos de los fármacos , Tibia/metabolismo , Tibia/patología
8.
Mol Cell Biol ; 34(11): 2029-45, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24662053

RESUMEN

The retinoblastoma protein (pRB) is best known for regulating cell proliferation through E2F transcription factors. In this report, we investigate the properties of a targeted mutation that disrupts pRB interactions with the transactivation domain of E2Fs. Mice that carry this mutation endogenously (Rb1(ΔG)) are defective for pRB-dependent repression of E2F target genes. Except for an accelerated entry into S phase in response to serum stimulation, cell cycle regulation in Rb1(ΔG/ΔG) mouse embryonic fibroblasts (MEFs) strongly resembles that of the wild type. In a serum deprivation-induced cell cycle exit, Rb1(ΔG/ΔG) MEFs display a magnitude of E2F target gene derepression similar to that of Rb1(-/-) cells, even though Rb1(ΔG/ΔG) cells exit the cell cycle normally. Interestingly, cell cycle arrest in Rb1(ΔG/ΔG) MEFs is responsive to p16 expression and gamma irradiation, indicating that alternate mechanisms can be activated in G1 to arrest proliferation. Some Rb1(ΔG/ΔG) mice die neonatally with a muscle degeneration phenotype, while the others live a normal life span with no evidence of spontaneous tumor formation. Most tissues appear histologically normal while being accompanied by derepression of pRB-regulated E2F targets. This suggests that non-E2F-, pRB-dependent pathways may have a more relevant role in proliferative control than previously identified.


Asunto(s)
Factores de Transcripción E2F/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/genética , Adenocarcinoma/genética , Alelos , Animales , Sitios de Unión , Línea Celular , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Fibroblastos/citología , Marcación de Gen , Ratones , Ratones Noqueados , Mutación , Neoplasias Hipofisarias/genética , Fase S/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA