Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Neurosci ; 17: 1289966, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38161998

RESUMEN

The tropomyosin receptor kinase B (TrkB) is encoded by the NTRK2 gene. It belongs to the family of transmembrane tyrosine kinases, which have key roles in the development and maintenance of the nervous system. Brain-derived neurotrophic factor (BDNF) and the neurotrophins NT3 and NT4/5 have high affinity for TrkB. Dysregulation of TrkB is associated to a large spectrum of diseases including neurodegeneration, psychiatric diseases and some cancers. The function of TrkB and its role in neural development have mainly been decrypted using transgenic mouse models, pharmacological modulators and human neuronal cell lines overexpressing NTRK2. In this study, we identified high expression and robust activity of TrkB in ReNcell VM, an immortalized human neural progenitor stem cell line and generated NTRK2-deficient (NTRK2-/-) ReNcell VM using the CRISPR/Cas9 gene editing technology. Global transcriptomic analysis revealed major changes in expression of specific genes responsible for neurogenesis, neuronal development and glial differentiation. In particular, key neurogenic transcription factors were massively down-regulated in NTRK2-/- cells, while early glial progenitor markers were enriched in NTRK2-/- cells compared to NTRK2+/+. This indicates a previously undescribed inhibitory role of TrkB on glial differentiation in addition to its well-described pro-neurogenesis role. Altogether, we have generated for the first time a human neural cell line with a loss-of-function mutation of NTRK2, which represents a reproducible and readily available cell culture system to study the role of TrkB during human neural differentiation, analyze the role of TrkB isoforms as well as validate TrkB antibodies and pharmacological agents targeting the TrkB pathway.

3.
Cancer Res ; 77(1): 62-73, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27815388

RESUMEN

Chromosome segregation during mitosis is monitored by the mitotic checkpoint and is dependent upon DNA methylation. ZBTB4 is a mammalian epigenetic regulator with high affinity for methylated CpGs that localizes at pericentromeric heterochromatin and is frequently downregulated in cancer. Here, we report that decreased ZBTB4 expression correlates with high genome instability across many frequent human cancers. In human cell lines, ZBTB4 depletion was sufficient to increase the prevalence of micronuclei and binucleated cells in parallel with aberrant mitotic checkpoint gene expression, a weakened mitotic checkpoint, and an increased frequency of lagging chromosomes during mitosis. To extend these findings, we generated Zbtb4-deficient mice. Zbtb4-/- mice were smaller than their wild-type littermates. Primary cells isolated from Zbtb4-/- mice exhibited diminished mitotic checkpoint activity, increased mitotic defects, aneuploid cells marked by a specific transcriptional signature, and increased genomic instability. Zbtb4-/- mice were also more susceptible to 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate (DMBA/TPA)-induced skin carcinogenesis. Our results establish the epigenetic regulator ZBTB4 as an essential component in maintaining genomic stability in mammals. Cancer Res; 77(1); 62-73. ©2016 AACR.


Asunto(s)
Aneuploidia , Transformación Celular Neoplásica/genética , Inestabilidad Genómica/genética , Puntos de Control de la Fase M del Ciclo Celular/genética , Neoplasias/genética , Proteínas Represoras/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Hibridación Fluorescente in Situ , Ratones , Ratones Noqueados , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Cutáneas/genética
4.
J Clin Endocrinol Metab ; 101(2): 513-22, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26695862

RESUMEN

CONTEXT: Cushing disease (CD) is due to pituitary corticotrope adenomas that produce unrestrained ACTH secretion and have lost the negative feedback exerted by glucocorticoids (GCs). GCs also restrain corticotrope proliferation, and the mechanisms of this inhibition are poorly understood. OBJECTIVE: The aim of the study was to identify cell cycle regulatory genes that are regulated by GCs and the glucocorticoid receptor and to assess regulatory genes that have a rate-limiting action on corticotrope proliferation and may be disregulated in CD. DESIGN: The mouse corticotrope tumor cells AtT-20 were used to identify GC-regulated genes that contribute to control of cell cycle progression. Surgery sections from patients with CD were used to assess expression of CABLES1 in corticotrope adenomas. METHODS: Gene expression profiling, small interfering RNA knockdowns, cell cycle analyses, and genetic manipulations were performed in AtT-20 cells. Sequencing of chromatin immunoprecipitation for pituitary-restricted transcription factors and RNA polymerase II were used to identify regulatory elements and genes that bind GR and are direct transcriptional targets. A panel of previously well-characterized corticotrope adenomas was used to correlate expression of CABLES1 with that of other markers. RESULTS: GCs altered expression of 3 positive and 3 negative regulators of cell cycle progression. Two Myc genes (L-Myc and N-Myc) and E2F2 are repressed by GCs, whereas genes for the negative regulators of the cell cycle, Gadd45ß, Gadd45γ, and Cables1 are activated by GCs. Cables1 small interfering RNA knockdown strongly stimulates AtT-20 cell proliferation and antagonizes the growth inhibition produced by GCs. The Gadd45 and Cables1 genes have the hallmarks of direct GC targets. CABLES1 is expressed in normal human pituitary cells, but expression is lost in ∼55% of corticotrope adenomas, and this is strongly correlated with the loss of p27(Kip1) expression. CONCLUSIONS: CABLES1 is a critical regulator of corticotrope proliferation that defines a pathway often inactivated in CD and links proliferation to GC resistance.


Asunto(s)
Hormona Adrenocorticotrópica/metabolismo , Proteínas Portadoras/genética , Ciclinas/genética , Glucocorticoides/metabolismo , Fosfoproteínas/genética , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/genética , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/metabolismo , Hipófisis/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Factor de Transcripción E2F2/genética , Regulación de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Genes cdc/genética , Genes myc/genética , Humanos , Ratones , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/patología , ARN Interferente Pequeño , Receptores de Glucocorticoides/genética
5.
Mol Endocrinol ; 24(9): 1835-45, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20660298

RESUMEN

Cushing's disease is caused by glucocorticoid-resistant pituitary corticotroph adenomas. We have previously identified the loss of nuclear Brg1 as one mechanism that may lead to partial glucocorticoid resistance: this loss is observed in about 33% of human corticotroph adenomas. We now show that Brg1 loss of function correlates with cyclin E expression in corticotroph adenomas and with loss of the cell cycle inhibitor p27(Kip1) expression. Because Brg1 is thought to have tumor suppressor activity, the present study was undertaken to understand the putative contribution of cyclin E derepression produced by loss of Brg1 expression on adenoma development. Overexpression of cyclin E in pituitary proopiomelanocortin cells leads to abnormal reentry into cell cycle of differentiated proopiomelanocortin cells and to centrosome instability. These alterations are consistent with the intermediate lobe hyperplasia and anterior lobe adenomas that were observed in these pituitaries. When combined with the p27(Kip1) knockout, overexpression of cyclin E increased the incidence of pituitary tumors, their size, and their proliferation index. These results suggest that cyclin E up-regulation and p27(Kip1) loss-of-function act cooperatively on pituitary adenoma development.


Asunto(s)
Ciclina E/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias Hipofisarias/metabolismo , Lesiones Precancerosas/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Proliferación Celular , Centrosoma/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , ADN Helicasas/metabolismo , Técnicas de Silenciamiento del Gen , Hiperplasia , Ratones , Ratones Transgénicos , Proteínas Nucleares/metabolismo , Fenotipo , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/complicaciones , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/metabolismo , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/patología , Hipófisis/metabolismo , Hipófisis/patología , Neoplasias Hipofisarias/complicaciones , Neoplasias Hipofisarias/patología , Lesiones Precancerosas/patología , Proopiomelanocortina/metabolismo , Proteínas Represoras , Factores de Transcripción/metabolismo
6.
Front Horm Res ; 38: 15-24, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20616491

RESUMEN

As model of organogenesis, the pituitary gland is a relatively simple tissue; yet, we understand little of the mechanisms that determine organ size, cell number and allocation of cells to different lineages. While the discovery of cell-restricted transcription factors has led to significant insight into the mechanisms controlling differentiation and cell-specific gene expression, we still need to integrate these processes with control of organ development. The identification of pituitary stem cells has suggested mechanisms for maintenance of adult pituitary but these findings again highlight the crucial role of cell cycle control for determination of progenitor and differentiated cell numbers. We recently described the mechanisms for progenitor cell cycle exit in early pituitary development that critically depend on the cell cycle inhibitor p57Kip2. It appears that cell cycle control is independent of differentiation, indicating that separate regulatory mechanisms must be involved in each process. The role of p57Kip2 appears to be restricted to progenitor cell cycle exit and it is rather the related p27Kip1 that prevents re-entry into the cycle of differentiated cells. While these data revealed a new transient intermediate between progenitors and differentiated cells, they also raised new questions and suggested that separate signals may control differentiation and cell cycle.


Asunto(s)
Ciclo Celular , Diferenciación Celular , Hipófisis/embriología , Células Madre/citología , Animales , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Proteínas de Homeodominio/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de Dominio T Box/fisiología
7.
Mol Cell Biol ; 29(7): 1895-908, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19139274

RESUMEN

Patterning and differentiation signals are often believed to drive the developmental program, including cell cycle exit of proliferating progenitors. Taking advantage of the spatial and temporal separation of proliferating and differentiated cells within the developing anterior pituitary gland, we investigated the control of cell proliferation during organogenesis. Thus, we identified a population of noncycling precursors that are uniquely marked by expression of the cell cycle inhibitor p57(Kip2) and by cyclin E. In p57(Kip2-/-) mice, the developing pituitary is hyperplastic due to accumulation of proliferating progenitors, whereas overexpression of p57(Kip2) leads to hypoplasia. p57(Kip2)-dependent cell cycle exit is not required for differentiation, and conversely, blockade of cell differentiation, as achieved in Tpit(-/-) pituitaries, does not prevent cell cycle exit but rather leads to accumulation of p57(Kip2)-positive precursors. Upon differentiation, p57(Kip2) is replaced by p27(Kip1). Accordingly, proliferating differentiated cells are readily detected in p27(Kip1-/-) pituitaries but not in wild-type or p57(Kip2-/-) pituitaries. Strikingly, all cells of p57(Kip2-/-);p27(Kip1-/-) pituitaries are proliferative. Thus, during normal development, progenitor cell cycle exit is controlled by p57(Kip2) followed by p27(Kip1) in differentiated cells; these sequential actions, taken together with different pituitary outcomes of their loss of function, suggest hierarchical controls of the cell cycle that are independent of differentiation.


Asunto(s)
Ciclo Celular , Diferenciación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Hipófisis/citología , Células Madre/citología , Animales , Proliferación Celular , Ciclina E/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/deficiencia , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...