Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 80(16): 3215-3221, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32606000

RESUMEN

Type 2 diabetes, which is mainly linked to obesity, is associated with increased incidence of liver cancer. We have previously found that in various models of obesity/diabetes, hyperinsulinemia maintains heightened hepatic expression of cyclin D1, suggesting a plausible mechanism linking diabetes and liver cancer progression. Here we show that cyclin D1 is greatly elevated in human livers with diabetes and is among the most significantly upregulated genes in obese/diabetic liver tumors. Liver-specific cyclin D1 deficiency protected obese/diabetic mice against hepatic tumorigenesis, whereas lean/nondiabetic mice developed tumors irrespective of cyclin D1 status. Cyclin D1 dependency positively correlated with liver cancer sensitivity to palbociclib, an FDA-approved CDK4 inhibitor, which was effective in treating orthotopic liver tumors under obese/diabetic conditions. The antidiabetic drug metformin suppressed insulin-induced hepatic cyclin D1 expression and protected against obese/diabetic hepatocarcinogenesis. These results indicate that the cyclin D1-CDK4 complex represents a potential selective therapeutic vulnerability for liver tumors in obese/diabetic patients. SIGNIFICANCE: Obesity/diabetes-associated liver tumors are specifically vulnerable to cyclin D1 deficiency and CDK4 inhibition, suggesting that the obese/diabetic environment confers cancer-selective dependencies that can be therapeutically exploited.


Asunto(s)
Ciclina D1/deficiencia , Diabetes Mellitus Tipo 2/complicaciones , Neoplasias Hepáticas Experimentales/etiología , Obesidad/complicaciones , Animales , Antineoplásicos/farmacología , Ciclina D1/genética , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Hiperinsulinismo/metabolismo , Hipoglucemiantes/farmacología , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/prevención & control , Masculino , Metformina/farmacología , Ratones , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Regulación hacia Arriba
2.
Nat Commun ; 10(1): 558, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30718512

RESUMEN

Inactivating mutations in SMARCA4 (BRG1), a key SWI/SNF chromatin remodelling gene, underlie small cell carcinoma of the ovary, hypercalcemic type (SCCOHT). To reveal its druggable vulnerabilities, we perform kinase-focused RNAi screens and uncover that SMARCA4-deficient SCCOHT cells are highly sensitive to the inhibition of cyclin-dependent kinase 4/6 (CDK4/6). SMARCA4 loss causes profound downregulation of cyclin D1, which limits CDK4/6 kinase activity in SCCOHT cells and leads to in vitro and in vivo susceptibility to CDK4/6 inhibitors. SCCOHT patient tumors are deficient in cyclin D1 yet retain the retinoblastoma-proficient/p16INK4a-deficient profile associated with positive responses to CDK4/6 inhibitors. Thus, our findings indicate that CDK4/6 inhibitors, approved for a breast cancer subtype addicted to CDK4/6 activation, could be repurposed to treat SCCOHT. Moreover, our study suggests a novel paradigm whereby critically low oncogene levels, caused by loss of a driver tumor suppressor, may also be exploited therapeutically.


Asunto(s)
Carcinoma de Células Pequeñas/tratamiento farmacológico , Carcinoma de Células Pequeñas/metabolismo , Ciclina D1/deficiencia , ADN Helicasas/metabolismo , Proteínas Nucleares/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Factores de Transcripción/metabolismo , Aminopiridinas/uso terapéutico , Animales , Bencimidazoles/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Inmunoprecipitación de Cromatina , Ciclina D1/metabolismo , ADN Helicasas/genética , Femenino , Humanos , Hipercalcemia/tratamiento farmacológico , Hipercalcemia/metabolismo , Ratones , Ratones SCID , Proteínas Nucleares/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Piperazinas/uso terapéutico , Purinas/uso terapéutico , Piridinas/uso terapéutico , ARN Interferente Pequeño/genética , Factores de Transcripción/genética
3.
Neural Dev ; 13(1): 23, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30219109

RESUMEN

BACKGROUND: Ex vivo, whole-mount explant culture of the rodent retina has proved to be a valuable approach for studying retinal development. In a limited number of recent studies, this method has been coupled to live fluorescent microscopy with the goal of directly observing dynamic cellular events. However, retinal tissue thickness imposes significant technical limitations. To obtain 3-dimensional images with high quality axial resolution, investigators are restricted to specific areas of the retina and require microscopes, such as 2-photon, with a higher level of depth penetrance. Here, we report a retinal live imaging method that is more amenable to a wider array of imaging systems and does not compromise resolution of retinal cross-sectional area. RESULTS: Mouse retinal slice cultures were prepared and standard, inverted confocal microscopy was used to generate movies with high quality resolution of retinal cross-sections. To illustrate the ability of this method to capture discrete, physiologically relevant events during retinal development, we imaged the dynamics of the Fucci cell cycle reporter in both wild type and Cyclin D1 mutant retinal progenitor cells (RPCs) undergoing interkinetic nuclear migration (INM). Like previously reported for the zebrafish, mouse RPCs in G1 phase migrated stochastically and exhibited overall basal drift during development. In contrast, mouse RPCs in G2 phase displayed directed, apical migration toward the ventricular zone prior to mitosis. We also determined that Cyclin D1 knockout RPCs in G2 exhibited a slower apical velocity as compared to wild type. These data are consistent with previous IdU/BrdU window labeling experiments on Cyclin D1 knockout RPCs indicating an elongated cell cycle. Finally, to illustrate the ability to monitor retinal neuron differentiation, we imaged early postnatal horizontal cells (HCs). Time lapse movies uncovered specific HC neurite dynamics consistent with previously published data showing an instructive role for transient vertical neurites in HC mosaic formation. CONCLUSIONS: We have detailed a straightforward method to image mouse retinal slice culture preparations that, due to its relative ease, extends live retinal imaging capabilities to a more diverse group of scientists. We have also shown that, by using a slice technique, we can achieve excellent lateral resolution, which is advantageous for capturing intracellular dynamics and overall cell movements during retinal development and differentiation.


Asunto(s)
Diagnóstico por Imagen/métodos , Retina/citología , Retina/diagnóstico por imagen , Retina/crecimiento & desarrollo , Factores de Edad , Animales , Animales Recién Nacidos , Calbindinas/metabolismo , Muerte Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Conexinas/genética , Conexinas/metabolismo , Ciclina D1/deficiencia , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histonas/metabolismo , Técnicas In Vitro , Ratones , Ratones Transgénicos , Componente 6 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Neurogénesis , Técnicas de Cultivo de Órganos , Factores de Tiempo
4.
J Cell Sci ; 131(12)2018 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-29880532

RESUMEN

Expression of cyclin D1 (CCND1) is required for cancer cell survival and proliferation. This is presumably due to the role of cyclin D1 in inactivation of the RB tumor suppressor. Here, we investigated the pro-survival function of cyclin D1 in a number of cancer cell lines. We found that cyclin D1 depletion facilitated cellular senescence in several cancer cell lines. Senescence triggered by cyclin D1 depletion was more extensive than that caused by the prolonged CDK4 inhibition. Intriguingly, the senescence caused by cyclin D1 depletion was independent of RB status of the cancer cell. We identified a build-up of intracellular reactive oxygen species in the cancer cells that underwent senescence upon depletion of cyclin D1 but not in those cells where CDK4 was inhibited. The higher ROS levels were responsible for the cell senescence, which was instigated by the p38-JNK-FOXO3a-p27 pathway. Therefore, expression of cyclin D1 prevents cancer cells from undergoing senescence, at least partially, by keeping the level of intracellular oxidative stress at a tolerable sub-lethal level. Depletion of cyclin D1 promotes the RB-independent pro-senescence pathway and the cancer cells then succumb to the endogenous oxidative stress levels.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Ciclina D1/deficiencia , Neoplasias/metabolismo , Neoplasias/patología , Estrés Oxidativo/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Senescencia Celular/fisiología , Ciclina D1/metabolismo , Humanos , Células MCF-7 , Proteína de Retinoblastoma/metabolismo
5.
Curr Drug Deliv ; 14(2): 246-252, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27174173

RESUMEN

BACKGROUND: Short interfering RNAs (siRNAs) are double-stranded RNA molecules able to specifically targeting genes products responsible for human diseases. Cyclin D1 (CyD1) is a cell cycleregulatory molecule, up-regulated at sites of inflammation in several tissues. CyD1 is a very interesting potential target in lung and colon inflammatory diseases. OBJECTIVE: The aim of this paper was testing CyD1 expression in human lung and colon tissues after the application of an inflammatory stimulus, and verifying its gene silencing by using siRNA for CyD1 (siCyD1). METHOD: Colon and pulmonary biopsies were treated with siCyD1 by using two different transfection carriers: a) invivofectamine and b) ad hoc produced nanoliposomes. After 24 hours of incubation with nanoliposomes encapsulating siRNA or invivofectamine-CyD1siRNA, in presence or absence of ECLPS, we analysed the protein expression of CyD1 through Western-Blotting. RESULTS: After EC-LPS treatment, in both colon and pulmonary biopsies, an overexpression of CyD1was found (about 64% and 40% respectively). Invivofectamine-CyD1 siRNA reduced the expression of CyD1 approximately by 46% compared to the basal condition, and by around 65% compared to EC-LPS treated colon samples. In lung, following in vivo fectamine siRNA silencing in the presence of EC-LPS, no reduction was observed. Ad hoc nanoliposomes were able to enter colon and lung tissues, but CyD1 silencing was reported in 2 colon samples out of 4 and no efficacy was demonstrated in the only lung sample we studied. CONCLUSION: The silencing of Cyclin D1 expression in vitro "organ culture" model is possible. Our preliminary results encourage further investigations, using different siRNA concentrations delivered by nanoliposomes.


Asunto(s)
Ciclina D1/deficiencia , Ciclina D1/genética , Silenciador del Gen , ARN Interferente Pequeño/genética , Técnicas de Cultivo de Tejidos , Femenino , Humanos , Masculino
6.
Nat Commun ; 7: 11581, 2016 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-27181366

RESUMEN

Cyclin D1 (Ccnd1) together with its binding partner Cdk4 act as a transcriptional regulator to control cell proliferation and migration, and abnormal Ccnd1·Cdk4 expression promotes tumour growth and metastasis. While different nuclear Ccnd1·Cdk4 targets participating in cell proliferation and tissue development have been identified, little is known about how Ccnd1·Cdk4 controls cell adherence and invasion. Here, we show that the focal adhesion component paxillin is a cytoplasmic substrate of Ccnd1·Cdk4. This complex phosphorylates a fraction of paxillin specifically associated to the cell membrane, and promotes Rac1 activation, thereby triggering membrane ruffling and cell invasion in both normal fibroblasts and tumour cells. Our results demonstrate that localization of Ccnd1·Cdk4 to the cytoplasm does not simply act to restrain cell proliferation, but constitutes a functionally relevant mechanism operating under normal and pathological conditions to control cell adhesion, migration and metastasis through activation of a Ccnd1·Cdk4-paxillin-Rac1 axis.


Asunto(s)
Ciclina D1/metabolismo , Citoplasma/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Paxillin/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Ciclina D1/deficiencia , Quinasa 4 Dependiente de la Ciclina/metabolismo , Regulación hacia Abajo/genética , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Ratas , Especificidad por Sustrato , Proteína de Unión al GTP rac1/metabolismo
7.
EMBO Rep ; 16(8): 1037-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26136374

RESUMEN

Although the two catalytic subunits of the SWI/SNF chromatin-remodeling complex--Brahma (Brm) and Brg1--are almost invariably co-expressed, their mutually exclusive incorporation into distinct SWI/SNF complexes predicts that Brg1- and Brm-based SWI/SNF complexes execute specific functions. Here, we show that Brg1 and Brm have distinct functions at discrete stages of muscle differentiation. While Brg1 is required for the activation of muscle gene transcription at early stages of differentiation, Brm is required for Ccnd1 repression and cell cycle arrest prior to the activation of muscle genes. Ccnd1 knockdown rescues the ability to exit the cell cycle in Brm-deficient myoblasts, but does not recover terminal differentiation, revealing a previously unrecognized role of Brm in the activation of late muscle gene expression independent from the control of cell cycle. Consistently, Brm null mice displayed impaired muscle regeneration after injury, with aberrant proliferation of satellite cells and delayed formation of new myofibers. These data reveal stage-specific roles of Brm during skeletal myogenesis, via formation of repressive and activatory SWI/SNF complexes.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , ADN Helicasas/metabolismo , Expresión Génica , Desarrollo de Músculos/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Ciclina D1/deficiencia , Ciclina D1/genética , ADN Helicasas/genética , Técnicas de Silenciamiento del Gen , Ratones , Células Musculares , Proteínas Nucleares/genética , Factores de Transcripción/genética
8.
Oncotarget ; 6(11): 8525-38, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25940700

RESUMEN

Cyclin D1 is an important molecular driver of human breast cancer but better understanding of its oncogenic mechanisms is needed, especially to enhance efforts in targeted therapeutics. Currently, pharmaceutical initiatives to inhibit cyclin D1 are focused on the catalytic component since the transforming capacity is thought to reside in the cyclin D1/CDK activity. We initiated the following study to directly test the oncogenic potential of catalytically inactive cyclin D1 in an in vivo mouse model that is relevant to breast cancer. Herein, transduction of cyclin D1(-/-) mouse embryonic fibroblasts (MEFs) with the kinase dead KE mutant of cyclin D1 led to aneuploidy, abnormalities in mitotic spindle formation, autosome amplification, and chromosomal instability (CIN) by gene expression profiling. Acute transgenic expression of either cyclin D1(WT) or cyclin D1(KE) in the mammary gland was sufficient to induce a high CIN score within 7 days. Sustained expression of cyclin D1(KE) induced mammary adenocarcinoma with similar kinetics to that of the wild-type cyclin D1. ChIP-Seq studies demonstrated recruitment of cyclin D1(WT) and cyclin D1(KE) to the genes governing CIN. We conclude that the CDK-activating function of cyclin D1 is not necessary to induce either chromosomal instability or mammary tumorigenesis.


Asunto(s)
Adenocarcinoma/genética , Ciclina D1/fisiología , Neoplasias Mamarias Experimentales/genética , Sustitución de Aminoácidos , Aneuploidia , Animales , Dominio Catalítico/genética , Transformación Celular Neoplásica/genética , Células Cultivadas , Centrosoma/ultraestructura , Inestabilidad Cromosómica/genética , Ciclina D1/deficiencia , Ciclina D1/genética , Femenino , Fibroblastos , Genes bcl-1 , Humanos , Virus del Tumor Mamario del Ratón/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Piperazinas/farmacología , Piridinas/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Huso Acromático/ultraestructura , Transducción Genética
9.
Nature ; 510(7506): 547-51, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24870244

RESUMEN

Insulin constitutes a principal evolutionarily conserved hormonal axis for maintaining glucose homeostasis; dysregulation of this axis causes diabetes. PGC-1α (peroxisome-proliferator-activated receptor-γ coactivator-1α) links insulin signalling to the expression of glucose and lipid metabolic genes. The histone acetyltransferase GCN5 (general control non-repressed protein 5) acetylates PGC-1α and suppresses its transcriptional activity, whereas sirtuin 1 deacetylates and activates PGC-1α. Although insulin is a mitogenic signal in proliferative cells, whether components of the cell cycle machinery contribute to its metabolic action is poorly understood. Here we report that in mice insulin activates cyclin D1-cyclin-dependent kinase 4 (Cdk4), which, in turn, increases GCN5 acetyltransferase activity and suppresses hepatic glucose production independently of cell cycle progression. Through a cell-based high-throughput chemical screen, we identify a Cdk4 inhibitor that potently decreases PGC-1α acetylation. Insulin/GSK-3ß (glycogen synthase kinase 3-beta) signalling induces cyclin D1 protein stability by sequestering cyclin D1 in the nucleus. In parallel, dietary amino acids increase hepatic cyclin D1 messenger RNA transcripts. Activated cyclin D1-Cdk4 kinase phosphorylates and activates GCN5, which then acetylates and inhibits PGC-1α activity on gluconeogenic genes. Loss of hepatic cyclin D1 results in increased gluconeogenesis and hyperglycaemia. In diabetic models, cyclin D1-Cdk4 is chronically elevated and refractory to fasting/feeding transitions; nevertheless further activation of this kinase normalizes glycaemia. Our findings show that insulin uses components of the cell cycle machinery in post-mitotic cells to control glucose homeostasis independently of cell division.


Asunto(s)
Ciclo Celular , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Transducción de Señal , Acetilación , Aminoácidos/farmacología , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Células Cultivadas , Ciclina D1/deficiencia , Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Diabetes Mellitus/metabolismo , Activación Enzimática , Ayuno , Eliminación de Gen , Gluconeogénesis/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Histona Acetiltransferasas/metabolismo , Homeostasis , Humanos , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo , Masculino , Ratones , Fosforilación , ARN Mensajero/análisis , ARN Mensajero/genética , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos
10.
Glia ; 61(9): 1443-55, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23839966

RESUMEN

Little is known about the molecular mechanisms driving proliferation of glial cells after an insult to the central nervous system (CNS). To test the hypothesis that the G1 regulator cyclin D1 is critical for injury-induced cell division of glial cells, we applied an injury model that causes brain damage within a well-defined region. For this, we injected the neurotoxin ibotenic acid into the prefrontal cortex of adult mice, which leads to a local nerve cell loss but does not affect the survival of glial cells. Here, we show that cyclin D1 immunoreativity increases drastically after neurotoxin injection. We find that the cyclin D1-immunopositive (cyclin D1+) cell population within the lesioned area consists to a large extent of Olig2+ oligodendrocyte progenitor cells. Analysis of cyclin D1-deficient mice demonstrates that the proliferation rate of Olig2+ cells diminishes upon loss of cyclin D1. Further, we show that cyclin-dependent kinase (cdk) 4, but not cdk6 or cdk2, is essential for driving cell division of Olig2-expressing cells in our injury model. These data suggest that distinct cell cycle proteins regulate proliferation of Olig2+ progenitor cells following a CNS insult.


Asunto(s)
Células Madre Adultas/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Lesiones Encefálicas/patología , Proliferación Celular , Corteza Cerebral/patología , Ciclina D1/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas del Tejido Nervioso/metabolismo , Análisis de Varianza , Animales , Bromodesoxiuridina , Proliferación Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Ciclina D1/deficiencia , Quinasa 2 Dependiente de la Ciclina/deficiencia , Quinasa 4 Dependiente de la Ciclina/deficiencia , Quinasa 6 Dependiente de la Ciclina/deficiencia , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ácido Iboténico/toxicidad , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Neurotoxinas/toxicidad , Factor de Transcripción 2 de los Oligodendrocitos , Factores de Tiempo
11.
Methods Mol Biol ; 942: 169-77, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23027051

RESUMEN

Short interfering RNAs (siRNAs) have become valued tools for knocking down specific genes. As such, siRNAs are routinely used to study gene function and are also being explored as therapeutic agents. Traditionally, siRNAs are designed to target one specific gene, but this chapter describes a procedure for designing dual-targeting siRNAs where the two strands in the siRNA duplex are both active and down-regulate different target genes through both siRNA and miRNA-like effects. The procedure can be used to create siRNAs that robustly target pairs of genes.


Asunto(s)
Diseño de Fármacos , MicroARNs/genética , ARN Bicatenario/química , ARN Bicatenario/genética , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , Secuencia de Bases , Ciclina D1/deficiencia , Ciclina D1/genética , Receptores ErbB/deficiencia , Receptores ErbB/genética , Estabilidad del ARN , ARN Mensajero/genética
12.
Am J Pathol ; 181(1): 294-302, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22658484

RESUMEN

Epidemiological and experimental studies have revealed an important role for prolactin (PRL) in breast cancer. Cyclin D1 is a major downstream target of PRL in lobuloalveolar development during pregnancy and is amplified and/or overexpressed in many breast carcinomas. To examine the importance of cyclin D1 in PRL-induced pathogenesis, we generated transgenic mice (NRL-PRL) that overexpress PRL in mammary epithelial cells, with wild-type, heterozygous, or genetically ablated cyclin D1 in the FVB/N genetic background. Although loss of one cyclin D1 allele did not affect PRL-induced mammary lesions in nonparous females, the complete absence of cyclin D1 (D1(-/-)) markedly decreased tumor incidence. Nevertheless, NRL-PRL/D1(-/-) females developed significantly more preneoplastic lesions (eg, epithelial hyperplasias and mammary intraepithelial neoplasias) than D1(-/-) females. Moreover, although lack of cyclin D1 reduced proliferation of morphologically normal mammary epithelium, transgenic PRL restored it to rates of wild-type females. PRL posttranscriptionally increased nuclear cyclin D3 protein in D1(-/-) luminal cells, indicating one compensatory mechanism. Consistently, pregnancy induced extensive lobuloalveolar growth in the absence of cyclin D1. However, transcripts for milk proteins were reduced, and pups failed to survive, suggesting that mammary differentiation was inadequate. Together, these results indicate that cyclin D1 is an important, but not essential, mediator of PRL-induced mammary proliferation and pathology in FVB/N mice and is critical for differentiation and lactation.


Asunto(s)
Adenocarcinoma/fisiopatología , Ciclina D1/fisiología , Neoplasias Mamarias Experimentales/fisiopatología , Prolactina/fisiología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ciclina D1/deficiencia , Ciclina D1/metabolismo , Ciclina D3/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Lactancia/fisiología , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/prevención & control , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiología , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Embarazo , Prolactina/metabolismo
14.
Nature ; 474(7350): 230-4, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21654808

RESUMEN

Cyclin D1 is a component of the core cell cycle machinery. Abnormally high levels of cyclin D1 are detected in many human cancer types. To elucidate the molecular functions of cyclin D1 in human cancers, we performed a proteomic screen for cyclin D1 protein partners in several types of human tumours. Analyses of cyclin D1 interactors revealed a network of DNA repair proteins, including RAD51, a recombinase that drives the homologous recombination process. We found that cyclin D1 directly binds RAD51, and that cyclin D1-RAD51 interaction is induced by radiation. Like RAD51, cyclin D1 is recruited to DNA damage sites in a BRCA2-dependent fashion. Reduction of cyclin D1 levels in human cancer cells impaired recruitment of RAD51 to damaged DNA, impeded the homologous recombination-mediated DNA repair, and increased sensitivity of cells to radiation in vitro and in vivo. This effect was seen in cancer cells lacking the retinoblastoma protein, which do not require D-cyclins for proliferation. These findings reveal an unexpected function of a core cell cycle protein in DNA repair and suggest that targeting cyclin D1 may be beneficial also in retinoblastoma-negative cancers which are currently thought to be unaffected by cyclin D1 inhibition.


Asunto(s)
Ciclina D1/metabolismo , Reparación del ADN , Neoplasias/metabolismo , Mapeo de Interacción de Proteínas , Recombinasa Rad51/metabolismo , Animales , Línea Celular Tumoral , Ensayo Cometa , Ciclina D1/deficiencia , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Células HeLa , Humanos , Ratones , Neoplasias/genética , Neoplasias/patología , Unión Proteica/efectos de la radiación , Radiación Ionizante , Recombinación Genética/genética , Proteína de Retinoblastoma/deficiencia
15.
Oncogene ; 29(32): 4543-54, 2010 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-20562911

RESUMEN

Members of the Notch family are involved in the development of breast cancer in animal models and in humans. In young transgenic mice, expressing intracellular activated Notch1 (N1(IC)) in mammary cells, we found that CD24(+) CD29(high) progenitor cells had enhanced survival, and were expanded through a cyclin D1-dependent pathway. This expansion positively correlated with the later cyclin D1-dependent formation of basal-like ductal tumors. This expanded population exhibited abnormal differentiation skewed toward the basal cells, showed signs of pre-malignancy (low PTEN/p53 and high c-myc) and contained stem cells with impaired self-renewal in vivo, and more numerous multipotent, ductal-restricted progenitors. Our data suggest that N1(IC) can favor transformation of progenitor cells early in life through a cyclin D1-dependent pathway.


Asunto(s)
Ciclina D1/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Células Madre Multipotentes/patología , Receptor Notch1/genética , Receptor Notch1/metabolismo , Animales , Apoptosis , Antígeno CD24/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica , Ciclina D1/deficiencia , Femenino , Regulación Neoplásica de la Expresión Génica , Integrina beta1/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Experimentales/metabolismo , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Transgénicos , Células Madre Multipotentes/metabolismo , Fosfohidrolasa PTEN/genética , Proteína p53 Supresora de Tumor/genética
16.
Nature ; 463(7279): 374-8, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-20090754

RESUMEN

Cyclin D1 belongs to the core cell cycle machinery, and it is frequently overexpressed in human cancers. The full repertoire of cyclin D1 functions in normal development and oncogenesis is unclear at present. Here we developed Flag- and haemagglutinin-tagged cyclin D1 knock-in mouse strains that allowed a high-throughput mass spectrometry approach to search for cyclin D1-binding proteins in different mouse organs. In addition to cell cycle partners, we observed several proteins involved in transcription. Genome-wide location analyses (chromatin immunoprecipitation coupled to DNA microarray; ChIP-chip) showed that during mouse development cyclin D1 occupies promoters of abundantly expressed genes. In particular, we found that in developing mouse retinas-an organ that critically requires cyclin D1 function-cyclin D1 binds the upstream regulatory region of the Notch1 gene, where it serves to recruit CREB binding protein (CBP) histone acetyltransferase. Genetic ablation of cyclin D1 resulted in decreased CBP recruitment, decreased histone acetylation of the Notch1 promoter region, and led to decreased levels of the Notch1 transcript and protein in cyclin D1-null (Ccnd1(-/-)) retinas. Transduction of an activated allele of Notch1 into Ccnd1(-/-) retinas increased proliferation of retinal progenitor cells, indicating that upregulation of Notch1 signalling alleviates the phenotype of cyclin D1-deficiency. These studies show that in addition to its well-established cell cycle roles, cyclin D1 has an in vivo transcriptional function in mouse development. Our approach, which we term 'genetic-proteomic', can be used to study the in vivo function of essentially any protein.


Asunto(s)
Ciclina D1/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteómica , Transcripción Genética , Alelos , Animales , Proteína de Unión a CREB/metabolismo , Inmunoprecipitación de Cromatina , Ciclina D1/deficiencia , Ciclina D1/genética , Genoma/genética , Ensayos Analíticos de Alto Rendimiento , Histona Acetiltransferasas/metabolismo , Espectrometría de Masas , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteómica/métodos , Ratas , Receptor Notch1/genética , Receptor Notch1/metabolismo , Retina/citología , Retina/embriología , Retina/metabolismo , Células Madre/citología , Células Madre/metabolismo
17.
Haematologica ; 94(11): 1555-62, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19880778

RESUMEN

BACKGROUND: Cyclin D1-negative mantle cell lymphoma is difficult to distinguish from other small B-cell lymphomas. The clinical and pathological characteristics of patients with this form of lymphoma have not been well defined. Overexpression of the transcription factor SOX11 has been observed in conventional mantle cell lymphoma. The aim of this study was to determine whether this gene is expressed in cyclin D1-negative mantle cell lymphoma and whether its detection may be useful to identify these tumors. DESIGN AND METHODS: The microarray database of 238 mature B-cell neoplasms was re-examined. SOX11 protein expression was investigated immunohistochemically in 12 cases of cyclin D1-negative mantle cell lymphoma, 54 cases of conventional mantle cell lymphoma, and 209 additional lymphoid neoplasms. RESULTS: SOX11 mRNA was highly expressed in conventional and cyclin D1-negative mantle cell lymphoma and in 33% of the cases of Burkitt's lymphoma but not in any other mature lymphoid neoplasm. SOX11 nuclear protein was detected in 50 cases (93%) of conventional mantle cell lymphoma and also in the 12 cyclin D1-negative cases of mantle cell lymphoma, the six cases of lymphoblastic lymphomas, in two of eight cases of Burkitt's lymphoma, and in two of three T-prolymphocytic leukemias but was negative in the remaining lymphoid neoplasms. Cyclin D2 and D3 mRNA levels were significantly higher in cyclin D1-negative mantle cell lymphoma than in conventional mantle cell lymphoma but the protein expression was not discriminative. The clinico-pathological features and outcomes of the patients with cyclin D1-negative mantle cell lymphoma identified by SOX11 expression were similar to those of patients with conventional mantle cell lymphoma. CONCLUSIONS: SOX11 mRNA and nuclear protein expression is a highly specific marker for both cyclin D1-positive and negative mantle cell lymphoma.


Asunto(s)
Ciclina D1/deficiencia , Linfoma de Células del Manto/patología , Factores de Transcripción SOXC/análisis , Adulto , Anciano , Biomarcadores de Tumor , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma/patología , Linfoma de Células del Manto/química , Linfoma de Células del Manto/terapia , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/genética , ARN Mensajero/análisis , Factores de Transcripción SOXC/genética , Resultado del Tratamiento
18.
Neural Dev ; 4: 15, 2009 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-19416500

RESUMEN

BACKGROUND: Maintaining the correct balance of proliferation versus differentiation in retinal progenitor cells (RPCs) is essential for proper development of the retina. The cell cycle regulator cyclin D1 is expressed in RPCs, and mice with a targeted null allele at the cyclin D1 locus (Ccnd1-/-) have microphthalmia and hypocellular retinas, the latter phenotype attributed to reduced RPC proliferation and increased photoreceptor cell death during the postnatal period. How cyclin D1 influences RPC behavior, especially during the embryonic period, is unclear. RESULTS: In this study, we show that embryonic RPCs lacking cyclin D1 progress through the cell cycle at a slower rate and exit the cell cycle at a faster rate. Consistent with enhanced cell cycle exit, the relative proportions of cell types born in the embryonic period, such as retinal ganglion cells and photoreceptor cells, are increased. Unexpectedly, cyclin D1 deficiency decreases the proportions of other early born retinal neurons, namely horizontal cells and specific amacrine cell types. We also found that the laminar positioning of horizontal cells and other cell types is altered in the absence of cyclin D1. Genetically replacing cyclin D1 with cyclin D2 is not efficient at correcting the phenotypes due to the cyclin D1 deficiency, which suggests the D-cyclins are not fully redundant. Replacement with cyclin E or inactivation of cyclin-dependent kinase inhibitor p27Kip1 restores the balance of RPCs and retinal cell types to more normal distributions, which suggests that regulation of the retinoblastoma pathway is an important function for cyclin D1 during embryonic retinal development. CONCLUSION: Our findings show that cyclin D1 has important roles in RPC cell cycle regulation and retinal histogenesis. The reduction in the RPC population due to a longer cell cycle time and to an enhanced rate of cell cycle exit are likely to be the primary factors driving retinal hypocellularity and altered output of precursor populations in the embryonic Ccnd1-/- retina.


Asunto(s)
Ciclina D1/metabolismo , Células Madre Embrionarias/fisiología , Neurogénesis/fisiología , Retina/citología , Retina/embriología , Animales , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Ciclo Celular/genética , Ciclina D1/deficiencia , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Noqueados , Neuronas/fisiología , Factores de Transcripción Otx/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Retina/crecimiento & desarrollo , Timidina/metabolismo , Factor de Transcripción Brn-3B/metabolismo , Tubulina (Proteína)/metabolismo
19.
Cell Death Differ ; 16(6): 869-78, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19229248

RESUMEN

Pifithrin-alpha (PFT-alpha) was shown to specifically block transcriptional activity of the tumor suppressor p53 and was therefore proposed to be useful in preventing the severe side effects often associated with chemo- and radiotherapy. We report here that although PFT-alpha efficiently protected different cell types from DNA damage-induced apoptosis, it mediated this effect regardless of the presence or absence of p53. Interestingly, PFT-alpha blocked the apoptosome-mediated processing and activation of caspase-9 and -3 without interfering with the activation of mitochondria. Neither the DNA damage-induced activation of Bax or Bak nor the loss of the mitochondrial membrane potential or the final release of cytochrome c were inhibited by this compound. Instead, the ability of PFT-alpha to protect p53-deficient cells from DNA damage-induced caspase activation and apoptosis was greatly diminished after siRNA-mediated downregulation of cyclin-D1 expression. In contrast, downregulation of other proteins involved in cell-cycle progression, such as the retinoblastoma protein, cyclin D3, as well as the cyclin-dependent kinases, 2, 4 and 6, could not abolish this protection. Thus, our data show that PFT-alpha protects cells from DNA damage-induced apoptosis also by a p53-independent mechanism that takes place downstream of mitochondria and that might involve cyclin D1.


Asunto(s)
Apoptosis , Benzotiazoles/farmacología , Daño del ADN , Mitocondrias/fisiología , Tolueno/análogos & derivados , Proteína p53 Supresora de Tumor/metabolismo , Apoptosomas/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular , Ciclina D1/deficiencia , Ciclina D1/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Humanos , ARN Interferente Pequeño/metabolismo , Radiación Ionizante , Proteína de Retinoblastoma/metabolismo , Tolueno/farmacología , Proteína p53 Supresora de Tumor/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...