Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Nutr Food Res ; 63(2): e1801096, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30383332

RESUMEN

SCOPE: The tumor suppressor p107, a pocket protein member of the retinoblastoma susceptibility protein family, plays an important role in the cell cycle and cellular adipocyte differentiation. Nonetheless, the mechanism by which it influences whole body Energy homeostasis is unknown. METHODS AND RESULTS: The phenotype of p107 knockout (KO) mixed-background C57BL6/129 mice phenotype is studied by focusing on the involvement of white and brown adipose tissue (WAT and BAT) in energy metabolism. It is shown that p107 KO mice are leaner and have high-fat diet resistence. This phenomenon is explained by an increase of energy expenditure. The higher energy expenditure is caused by the activation of thermogenesis and may be mediated by both BAT and the browning of WAT. Consequently, it leads to the resistance of p107 KO mice to high-fat diet effects, prevention of liver steatosis, and improvement of the lipid profile and glucose homeostasis. CONCLUSION: These data allowed the unmasking of a mechanism by which a KO of p107 prevents diet-induced obesity by increasing energy expenditure via increased thermogenesis in BAT and browning of WAT, indicating the relevance of p107 as a modulator of metabolic activity of both brown and white adipocytes. Therefore, it can be targeted for the development of new therapies to ameliorate the metabolic syndrome.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Metabolismo Energético , Proteína p107 Similar a la del Retinoblastoma/fisiología , Termogénesis , Animales , Dieta Alta en Grasa , Hígado Graso/prevención & control , Glucosa/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína p107 Similar a la del Retinoblastoma/deficiencia
2.
Genes Chromosomes Cancer ; 56(3): 231-242, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27750399

RESUMEN

Several murine retinoblastoma models have been generated by deleting the genes encoding for retinoblastoma susceptibility protein pRb and one of its family members p107 or p130. In Rb-/- p107-/- retinoblastomas, somatic copy number alterations (SCNAs) like Mdm2 amplification or Cdkn2a deletion targeting the p53-pathway occur, which is uncommon for human retinoblastoma. In our study, we determined SCNAs in retinoblastomas developing in Rb-/- p130-/- mice and compared this to murine Rb-/- p107-/- tumors and human tumors. Chimeric mice were made by injection of 129/Ola-derived Rb-/- p130-/- embryonic stem cells into wild type C57BL/6 blastocysts. SCNAs of retinoblastoma samples were determined by low-coverage (∼0.5×) whole genome sequencing. In Rb-/- p130-/- tumors, SCNAs included gain of chromosomes 1 (3/23 tumors), 8 (1/23 tumors), 10 (1/23 tumors), 11 (2/23 tumors), and 12 (4/23 tumors), which could be mapped to frequently altered chromosomes in human retinoblastomas. While the altered chromosomes in Rb-/- p130-/- tumors were similar to those in Rb-/- p107-/- tumors, the alteration frequencies were much lower in Rb-/- p130-/- tumors. Most of the Rb-/- p130-/- tumors (16/23 tumors, 70%) were devoid of SCNAs, in strong contrast to Rb-/- p107-/- tumors, which were never (0/15 tumors) SCNA-devoid. Similarly, to human retinoblastoma, increased age at diagnosis significantly correlated with increased SCNA frequencies. Additionally, focal loss of Cdh11 was observed in one Rb-/- p130-/- tumor, which enforces studies in human retinoblastoma that identified CDH11 as a retinoblastoma suppressor. Moreover, based on a comparison of genes altered in human and murine retinoblastoma, we suggest exploring the role of HMGA1 and SRSF3 in retinoblastoma development. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Biomarcadores de Tumor/genética , Variaciones en el Número de Copia de ADN/genética , Proteína p107 Similar a la del Retinoblastoma/fisiología , Proteína p130 Similar a la del Retinoblastoma/fisiología , Retinoblastoma/genética , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
3.
J Vis Exp ; (95): 51983, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25650557

RESUMEN

Genetic deletion using the Cre-Lox system in transgenic mouse lines is a powerful tool used to study protein function. However, except in very specific Cre models, deletion of a protein throughout a tissue or cell population often leads to complex phenotypes resulting from multiple interacting mechanisms. Determining whether a phenotype results from disruption of a cell autonomous mechanism, which is intrinsic to the cell in question, or from a non-cell autonomous mechanism, which would result from impairment of that cell's environment, can be difficult to discern. To gain insight into protein function in an in vivo context, in utero electroporation (IUE) enables gene deletion in a small subset of cells within the developing cortex or some other selected brain region. IUE can be used to target specific brain areas, including the dorsal telencephalon, medial telencephalon, hippocampus, or ganglionic eminence. This facilitates observation of the consequences of cell autonomous gene deletion in the context of a healthy environment. The goal of this protocol is to show how IUE can be used to analyze a defect in radial migration in a floxed transgenic mouse line, with an emphasis on distinguishing between the cell autonomous and non-cell autonomous effects of protein deletion. By comparing the phenotype resulting from gene deletion within the entire cortex versus IUE-mediated gene deletion in a limited cell population, greater insight into protein function in brain development can be obtained than by using either technique in isolation.


Asunto(s)
Encéfalo/fisiología , Movimiento Celular/fisiología , Electroporación/métodos , Técnicas de Silenciamiento del Gen/métodos , Neuronas/fisiología , Proteína de Retinoblastoma/fisiología , Proteína p107 Similar a la del Retinoblastoma/fisiología , Animales , Encéfalo/citología , Embrión de Mamíferos , Femenino , Eliminación de Gen , Masculino , Ratones , Ratones Transgénicos , Neuronas/citología , Embarazo , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/genética
4.
Cell Death Differ ; 20(7): 931-40, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23558950

RESUMEN

The 'activating' E2fs (E2f1-3) are transcription factors that potently induce quiescent cells to divide. Work on cultured fibroblasts suggested they were essential for division, but in vivo analysis in the developing retina and other tissues disproved this notion. The retina, therefore, is an ideal location to assess other in vivo adenovirus E2 promoter binding factor (E2f) functions. It is thought that E2f1 directly induces apoptosis, whereas other activating E2fs only induce death indirectly by upregulating E2f1 expression. Indeed, mouse retinoblastoma (Rb)-null retinal neuron death requires E2f1, but not E2f2 or E2f3. However, we report an entirely distinct mechanism in dying cone photoreceptors. These neurons survive Rb loss, but undergo apoptosis in the cancer-prone retina lacking both Rb and its relative p107. We show that while E2f1 killed Rb/p107 null rod, bipolar and ganglion neurons, E2f2 was required and sufficient for cone death, independent of E2f1 and E2f3. Moreover, whereas E2f1-dependent apoptosis was p53 and p73-independent, E2f2 caused p53-dependent cone death. Our in vivo analysis of cone photoreceptors provides unequivocal proof that E2f-induces apoptosis independent of E2f1, and reveals distinct E2f1- and E2f2-activated death pathways in response to a single tumorigenic insult.


Asunto(s)
Apoptosis/fisiología , Factor de Transcripción E2F1/fisiología , Factor de Transcripción E2F2/fisiología , Factor de Transcripción E2F3/fisiología , Células Fotorreceptoras Retinianas Conos/patología , Animales , Apoptosis/genética , División Celular/genética , División Celular/fisiología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Factor de Transcripción E2F1/deficiencia , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F2/deficiencia , Factor de Transcripción E2F2/genética , Factor de Transcripción E2F3/deficiencia , Factor de Transcripción E2F3/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Retina/patología , Retina/fisiopatología , Células Fotorreceptoras Retinianas Conos/fisiología , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/fisiología , Proteína p107 Similar a la del Retinoblastoma/deficiencia , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/fisiología , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología
5.
Oncogene ; 32(40): 4798-805, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23146901

RESUMEN

The pocket proteins pRB, p107 and p130 have established roles in regulating the cell cycle through the control of E2F activity. The pocket proteins regulate differentiation of a number of tissues in both cell cycle-dependent and -independent manners. Prior studies showed that mutation of p107 and p130 in the mouse leads to defects in cartilage development during endochondral ossification, the process by which long bones form. Despite evidence of a role for pRB in osteoblast differentiation, it is unknown whether it functions during cartilage development. Here, we show that mutation of Rb in the early mesenchyme of p107-mutant mice results in severe cartilage defects in the growth plates of long bones. This is attributable to inappropriate chondrocyte proliferation that persists after birth and leads to the formation of enchondromas in the growth plates as early as 8 weeks of age. Genetic crosses show that development of these tumorigenic lesions is E2f3 dependent. These results reveal an overlapping role for pRB and p107 in cartilage development, endochondral ossification and enchondroma formation that reflects their coordination of cell-cycle exit at appropriate developmental stages.


Asunto(s)
Condrogénesis/fisiología , Condroma/genética , Placa de Crecimiento/crecimiento & desarrollo , Proteína de Retinoblastoma/fisiología , Proteína p107 Similar a la del Retinoblastoma/fisiología , Animales , Condrogénesis/genética , Condroma/patología , Ratones , Ratones Noqueados , Mutación , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/genética , Tomografía Computarizada por Rayos X
6.
Nat Commun ; 3: 1244, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23212373

RESUMEN

The mechanisms ensuring the long-term self-renewal of human embryonic stem cells are still only partly understood, limiting their use in cellular therapies. Here we found that increased activity of the RB cell cycle inhibitor in human embryonic stem cells induces cell cycle arrest, differentiation and cell death. Conversely, inactivation of the entire RB family (RB, p107 and p130) in human embryonic stem cells triggers G2/M arrest and cell death through functional activation of the p53 pathway and the cell cycle inhibitor p21. Differences in E2F target gene activation upon loss of RB family function between human embryonic stem cells, mouse embryonic stem cells and human fibroblasts underscore key differences in the cell cycle regulatory networks of human embryonic stem cells. Finally, loss of RB family function promotes genomic instability in both human and mouse embryonic stem cells, uncoupling cell cycle defects from chromosomal instability. These experiments indicate that a homeostatic level of RB activity is essential for the self-renewal and the survival of human embryonic stem cells.


Asunto(s)
Supervivencia Celular/fisiología , Células Madre Embrionarias/fisiología , Proteína de Retinoblastoma/fisiología , Animales , Ciclo Celular/fisiología , Muerte Celular/fisiología , Proteína Sustrato Asociada a CrK/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Fibroblastos/fisiología , Humanos , Ratones , Proteína p107 Similar a la del Retinoblastoma/fisiología , Proteína p53 Supresora de Tumor/fisiología
7.
Cancer Metastasis Rev ; 28(3-4): 281-90, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20012171

RESUMEN

Urothelium, one of the slowest cycling epithelia in the body, embodies a unique biological context for cellular transformation. Introduction of oncogenes into or removing tumor suppressor genes from the urothelial cells or a combination of both using the transgenic and/or knockout mouse approaches has provided useful insights into the molecular mechanisms of urothelial transformation and tumorigenesis. It is becoming increasingly clear that over-activation of the receptor tyrosine kinase (RTK) pathway, as exemplified by the constitutively activated Ha-ras oncogene, is both necessary and sufficient to initiate the low-grade, non-invasive urothelial carcinomas. Dosage of the mutated Ha-ras, but not concurrent inactivation of pro-senescence molecules p16Ink4a and p19Arf, dictates whether and when the low-grade urothelial carcinomas arise. Inactivation of both p53 and pRb, a prevailing paradigm previously proposed for muscle-invasive urothelial tumorigenesis, is found to be necessary but insufficient to initiate this urothelial carcinoma variant. Instead, downregulation in p53/pRb co-deficient urothelial cells of p107, a pRb family member, is associated with the genesis of the muscle-invasive bladder cancers. p53 deficiency also seems to be capable of cooperating with that of PTEN in eliciting invasive urothelial carcinomas. The genetically engineered mice have improved the molecular definition of the divergent pathways of urothelial tumorigenesis and progression, helped delineate the intricate crosstalk among different genetic alterations within a urothelium-specific context, identified new prognostic markers and novel therapeutic targets potentially applicable for clinical intervention, and provided in vivo platforms for testing preventive strategies of bladder cancer.


Asunto(s)
Carcinoma de Células Transicionales/genética , Transformación Celular Neoplásica/genética , Ratones Transgénicos/genética , Neoplasias Urológicas/genética , Animales , Carcinoma de Células Transicionales/diagnóstico , Carcinoma de Células Transicionales/patología , Genes de Retinoblastoma , Genes p53 , Genes ras , Ingeniería Genética , Humanos , Hiperplasia , Ratones , Ratones Noqueados , Invasividad Neoplásica/genética , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/fisiología , Neoplasias Urológicas/diagnóstico , Neoplasias Urológicas/patología , Urotelio/patología
8.
Diabetes ; 58(8): 1852-62, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19509021

RESUMEN

OBJECTIVE: The retinoblastoma protein family (pRb, p130, p107) plays a central role in the regulation of cell cycle progression. Surprisingly, loss of pRb in the beta-cell has no discernible effect on cell cycle control. Therefore, we explored the effects of individual loss of either p130 or p107 in addition to the simultaneous loss of both pRb/p130 on the beta-cell. RESEARCH DESIGN AND METHODS: Adult mice deficient in either p130 or p107 or both pRb/p130 were examined for effects on beta-cell replication, function, and survival. The Cre-Lox system was also used to inactivate pRb in wild-type and p130-deficient beta-cells in vitro. RESULTS: In vivo loss of either p107 or p130 did not affect beta-cell replication or function. Combined pRb/p130 loss, however, resulted in dramatically accelerated proliferation as well as apoptotic cell death. Pancreas and beta-cell mass were significantly reduced in double mutants. Despite this, overall glucose tolerance was normal, except for mild postprandial hyperglycemia. Ex vivo, acute deletion of pRb in p130-deficient beta-cells also caused a striking increase in proliferation. The combined deletion of pRb/p130 upregulated islet expression of E2F2 but not E2F1. CONCLUSIONS: These studies define an essential role for the pocket proteins in controlling the G(1)/S transition in beta-cells. When deficient in both pRb and p130, beta-cells undergo unrestrained cell cycle reentry and activation of apoptosis. These studies underscore the central role of the pRb pathway in controlling beta-cell turnover and provide new cellular targets for beta-cell regeneration.


Asunto(s)
Ciclo Celular/fisiología , Fase G1/fisiología , Células Secretoras de Insulina/citología , Proteína de Retinoblastoma/fisiología , Proteína p107 Similar a la del Retinoblastoma/fisiología , Proteína p130 Similar a la del Retinoblastoma/fisiología , Fase S/fisiología , Animales , Apoptosis/fisiología , Glucemia/metabolismo , División Celular , Prueba de Tolerancia a la Glucosa , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/deficiencia , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/deficiencia , Proteína p130 Similar a la del Retinoblastoma/genética
9.
Mol Carcinog ; 47(2): 105-13, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17932945

RESUMEN

The specific deletion of Rb gene in epidermis leads to altered proliferation and differentiation, but not to the development of spontaneous tumors. Our previous data have demonstrated the existence of a functional compensation of Rb loss by Rbl1 (p107) in as the phenotypic differences with respect to controls are intensified. However, the possible evolution of this aggravated phenotype, in particular in relationship with tumorigenesis, has not been evaluated due to the premature death of the double deficient mice. We have now investigated whether p107 can also act as a tumor suppressor in pRb-deficient epidermis using different experimental approaches. We found spontaneous tumor development in doubly-deficient skin grafts. Moreover, Rb-deficient keratinocytes are susceptible to Ha-ras-induced transformation, and this susceptibility is enhanced by p107 loss. Further functional analyses, including microarray gene expression profiling, indicated that the loss of p107, in the absence of pRb, produces the reduction of p53-dependent pro-apoptotic signals. Overall, our data demonstrate that p107 behaves as a tumor suppressor in epidermis in the absence of pRb and suggest novel tumor-suppressive roles for p107 in the context of functional p53 and activated Ras.


Asunto(s)
Genes de Retinoblastoma , Proteína p107 Similar a la del Retinoblastoma/fisiología , Neoplasias Cutáneas/genética , Piel/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Células Cultivadas , Femenino , Eliminación de Gen , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Desnudos , Proteína p107 Similar a la del Retinoblastoma/genética
10.
Cell ; 131(2): 378-90, 2007 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17956737

RESUMEN

During neurogenesis, the progression from a progenitor cell to a differentiated neuron is believed to be unidirectional and irreversible. The Rb family of proteins (Rb, p107, and p130) regulates cell-cycle exit and differentiation during retinogenesis. Rb and p130 are redundantly expressed in the neurons of the inner nuclear layer (INL) of the retina. We have found that in the adult Rb;p130-deficient retinae p107 compensation prevents ectopic proliferation of INL neurons. However, p107 is haploinsufficient in this process. Differentiated Rb(-/-);p107(+/-);p130(-/-) horizontal interneurons re-entered the cell cycle, clonally expanded, and formed metastatic retinoblastoma. Horizontal cells were not affected in Rb(+/-);p107(-/-);p130(-/-) or Rb(-/-);p107(-/-);p130(+/-), retinae suggesting that one copy of Rb or p130 was sufficient to prevent horizontal proliferation. We hereby report that differentiated neurons can proliferate and form cancer while maintaining their differentiated state including neurites and synaptic connections.


Asunto(s)
Interneuronas/fisiología , Neoplasias de la Retina/patología , Retinoblastoma/secundario , Células Madre/fisiología , Animales , Neoplasias de la Médula Ósea/patología , Neoplasias de la Médula Ósea/secundario , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Diferenciación Celular , División Celular , Interneuronas/metabolismo , Metástasis Linfática , Ratones , Retina/patología , Retinoblastoma/patología , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/fisiología , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/fisiología , Células Madre/metabolismo
11.
Genes Dev ; 21(1): 49-54, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17210787

RESUMEN

Genetic studies have demonstrated that Bmi1 promotes cell proliferation and stem cell self-renewal with a correlative decrease of p16(INK4a) expression. Here, we demonstrate that Polycomb genes EZH2 and BMI1 repress p16 expression in human and mouse primary cells, but not in cells deficient for pRB protein function. The p16 locus is H3K27-methylated and bound by BMI1, RING2, and SUZ12. Inactivation of pRB family proteins abolishes H3K27 methylation and disrupts BMI1, RING2, and SUZ12 binding to the p16 locus. These results suggest a model in which pRB proteins recruit PRC2 to trimethylate p16, priming the BMI1-containing PRC1L ubiquitin ligase complex to silence p16.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/antagonistas & inhibidores , Metilación de ADN , Silenciador del Gen , Histonas/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteína p107 Similar a la del Retinoblastoma/fisiología , Proteína p130 Similar a la del Retinoblastoma/fisiología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Humanos , Ratones , Ratones Noqueados , Proteínas de Neoplasias , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 1 , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
12.
Pigment Cell Res ; 19(4): 260-83, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16827746

RESUMEN

Melanoma is the most lethal of human skin cancers and its incidence is increasing worldwide [L.K. Dennis (1999). Arch. Dermatol. 135, 275; C. Garbe et al. (2000). Cancer 89, 1269]. Melanomas often metastasize early during the course of the disease and are then highly intractable to current therapeutic regimens [M.F. Demierre and G. Merlino (2004). Curr. Oncol. Rep. 6, 406]. Consequently, understanding the factors that maintain melanocyte homeostasis and prevent their neoplastic transformation into melanoma is of utmost interest from the perspective of therapeutic interdiction. This review will focus on the role of the pocket proteins (PPs), Rb1 (retinoblastoma protein), retinoblastoma-like 1 (Rbl1 also known as p107) and retinoblastoma-like 2 (Rbl2 also known as p130), in melanocyte homeostasis, with particular emphasis on their functions in the cell cycle and the DNA damage repair response. The potential mechanisms of PP deregulation in melanoma and the possibility of PP-independent pathways to melanoma development will also be considered. Finally, the role of the PP family in ultraviolet radiation (UVR)-induced melanoma and the precise contribution that each PP family member makes to melanocyte homeostasis will be discussed in the context of a number of genetically engineered mouse models.


Asunto(s)
Melanocitos/fisiología , Melanoma/fisiopatología , Proteína de Retinoblastoma/fisiología , Proteína p107 Similar a la del Retinoblastoma/fisiología , Proteína p130 Similar a la del Retinoblastoma/fisiología , Animales , Ciclo Celular/fisiología , Daño del ADN/efectos de la radiación , Homeostasis , Humanos , Ratones , Factor de Transcripción Asociado a Microftalmía/fisiología , Modelos Animales , Modelos Biológicos , Neoplasias Inducidas por Radiación , Transducción de Señal , Rayos Ultravioleta/efectos adversos
13.
Cancer Cell ; 8(6): 455-66, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16338659

RESUMEN

Primary mouse embryonic fibroblasts lacking expression of all three retinoblastoma protein family members (TKO MEFs) have lost the G1 restriction point. However, in the absence of mitogens these cells become highly sensitive to apoptosis. Here, we show that TKO MEFs that survive serum depletion pass G1 but completely arrest in G2. p21CIP1 and p27KIP1 inhibit Cyclin A-Cdk2 activity and sequester Cyclin B1-Cdk1 in inactive complexes in the nucleus. This response is alleviated by mitogen restimulation or inactivation of p53. Thus, our results disclose a cell cycle arrest mechanism in G2 that restricts the proliferative capacity of mitogen-deprived cells that have lost the G1 restriction point. The involvement of p53 provides a rationale for the synergism between loss of Rb and p53 in tumorigenesis.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Mitógenos/fisiología , Proteína de Retinoblastoma/fisiología , Proteína p107 Similar a la del Retinoblastoma/fisiología , Proteína p130 Similar a la del Retinoblastoma/fisiología , Animales , Apoptosis/fisiología , Proteína Quinasa CDC2/antagonistas & inhibidores , Proteína Quinasa CDC2/efectos de los fármacos , Proteína Quinasa CDC2/metabolismo , Ciclo Celular/genética , Ciclina A/antagonistas & inhibidores , Ciclina A/metabolismo , Ciclina B/efectos de los fármacos , Ciclina B/metabolismo , Ciclina B1 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/farmacología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/farmacología , Daño del ADN , Fibroblastos/citología , Fibroblastos/metabolismo , Fase G1/efectos de los fármacos , Fase G1/fisiología , Fase G2/efectos de los fármacos , Fase G2/fisiología , Ratones , Ratones Noqueados , Mitógenos/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/genética , Proteína p53 Supresora de Tumor/metabolismo
14.
Cancer Res ; 65(22): 10243-54, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16288012

RESUMEN

Because each change in the evolution of a cancer is predicated on the effects of previous events, a full understanding of selective changes and their effect on tumor progression can only be understood in the context of appropriate initiating events. Here, we define the effect of pRb function inactivation in prostate epithelium on both the initiation of prostate cancer and the establishment of selective pressures that lead to diminished Pten function and tumor evolution. Using genetically engineered mice, we show that inactivation of the pRb family proteins (Rb/p107/p130) induces epithelial proliferation and apoptosis and is sufficient to produce prostatic intraepithelial neoplasia (PIN) lesions. Over time, adenocarcinomas develop in all mice with no evidence of neuroendocrine tumors. Apoptosis is dependent on Pten function and not p53, unlike other epithelial cell types tested previously. Consequently, Pten hemizygosity reduces apoptosis by 50%, accelerating progression to adenocarcinomas with heterogeneous composition. Heterogeneity is associated with concurrent Pten haploinsufficiency and focal selective progression to complete Pten loss, which yields distinct tumor properties. Given that this analysis models the apparent timing of highly penetrant events in human prostate cancer, observed effects may recapitulate the natural evolution of prostate cancer development.


Asunto(s)
Adenocarcinoma/patología , Transformación Celular Neoplásica/patología , Neoplasias de la Próstata/patología , Proteína de Retinoblastoma/antagonistas & inhibidores , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Apoptosis/fisiología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Masculino , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/fisiología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Proteína de Retinoblastoma/fisiología , Proteína p107 Similar a la del Retinoblastoma/antagonistas & inhibidores , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Proteína p107 Similar a la del Retinoblastoma/fisiología , Proteína p130 Similar a la del Retinoblastoma/antagonistas & inhibidores , Proteína p130 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma/fisiología
15.
Front Biosci ; 3: d11-24, 1998 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9405335

RESUMEN

Pocket proteins, including the retinoblastoma susceptibility gene product (pRB) and the related proteins p107 and p130, function at cell cycle regulatory steps that link cyclin/CDK-integrated positive and negative growth signals with E2F transcription factor activity on genes required for cell cycle progression. Protein complex formation between pocket proteins and members of the E2F family of transcription factors determines whether E2F complexes act as transcriptional activators or repressors. Experimental work over the last few years indicates that individual pocket proteins interact with specific E2F members to regulate the transcription of certain genes under diverse cell growth conditions. Among these protein associations, p130-containing E2F complexes seem to be of particular importance in controlling gene transcription in quiescent and differentiating cells by repressing the transcription of a set of E2F-responsive genes. Once the cells are progressing through the G1 phase of the cell cycle, pocket protein-mediated regulation of E2F activity is assumed by pRB and p107. p130-mediated transcriptional regulation thus seems to prevent a gene expression program characteristic of dividing cells at the cell cycle exit and re-entrance transitions and in quiescent cells.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Factor de Transcripción E2F4/fisiología , Proteína p130 Similar a la del Retinoblastoma/fisiología , Animales , Diferenciación Celular/fisiología , Fase G1/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Fosforilación , Fase de Descanso del Ciclo Celular/fisiología , Proteína p107 Similar a la del Retinoblastoma/fisiología , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA