Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Neuroendocrinology ; 110(9-10): 797-804, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32126562

RESUMEN

Jakob Erdheim (1874-1937) first described craniopharyn-giomas (CPs) as "hypophyseal duct tumours" and postulated the existence of two tumour types based on their histological features: (1) an aggressive type showing similarities to adamantinomas (tumours of the jaw) and (2) a more benign form characterised by the presence of papillary structures. More than a century later, these initial observations have been confirmed; based on their distinct genetic, epigenetic, and histological features, the WHO classifies CPs into two types: adamantinomatous CPs (ACPs) and papillary CPs (PCPs). Considerable knowledge has been generated on the biology of CPs in the last 20 years. Mutations in CTNNB1 (encoding ß-catenin) are prevalent in ACP, whilst PCPs frequently harbour mutations in BRAF (p.BRAF-V600E). The consequence of these mutations is the activation of either the WNT/ß-catenin (ACP) or the MAPK/ERK (PCP) pathway. Murine models support a critical role for these mutations in tumour formation and have provided important insights into tumour pathogenesis, mostly in ACP. A critical role for cellular senescence has been uncovered in murine models of ACP with relevance to human tumours. Several gene profiling studies of human and murine ACP tumours have identified potential targetable pathways, and novel therapeutic agents are being used in clinical and pre-clinical research, in some cases with excellent results. In this review, we will present the accumulated knowledge on the biological features of these tumours and summarise how these advances are being translated into potential novel treatments.


Asunto(s)
Craneofaringioma , Neoplasias Hipofisarias , Animales , Craneofaringioma/tratamiento farmacológico , Craneofaringioma/genética , Craneofaringioma/metabolismo , Craneofaringioma/patología , Humanos , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología
2.
Proc Natl Acad Sci U S A ; 113(5): E548-57, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26764381

RESUMEN

Aberrant embryonic development of the hypothalamus and/or pituitary gland in humans results in congenital hypopituitarism (CH). Transcription factor 7-like 1 (TCF7L1), an important regulator of the WNT/ß-catenin signaling pathway, is expressed in the developing forebrain and pituitary gland, but its role during hypothalamo-pituitary (HP) axis formation or involvement in human CH remains elusive. Using a conditional genetic approach in the mouse, we first demonstrate that TCF7L1 is required in the prospective hypothalamus to maintain normal expression of the hypothalamic signals involved in the induction and subsequent expansion of Rathke's pouch progenitors. Next, we reveal that the function of TCF7L1 during HP axis development depends exclusively on the repressing activity of TCF7L1 and does not require its interaction with ß-catenin. Finally, we report the identification of two independent missense variants in human TCF7L1, p.R92P and p.R400Q, in a cohort of patients with forebrain and/or pituitary defects. We demonstrate that these variants exhibit reduced repressing activity in vitro and in vivo relative to wild-type TCF7L1. Together, our data provide support for a conserved molecular function of TCF7L1 as a transcriptional repressor during HP axis development in mammals and identify variants in this transcription factor that are likely to contribute to the etiology of CH.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Proteína 1 Similar al Factor de Transcripción 7/fisiología , Animales , Estudios de Cohortes , Humanos , Ratones , Hipófisis/anomalías , Hipófisis/metabolismo , Hipófisis/fisiopatología , Prosencéfalo/anomalías , Prosencéfalo/metabolismo
3.
Cell Mol Life Sci ; 70(20): 3739-52, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23397132

RESUMEN

Research from the last 15 years has provided a working model for how the anterior forebrain is induced and specified during the early stages of embryogenesis. This model relies on three basic processes: (1) induction of the neural plate from naive ectoderm requires the inhibition of BMP/TGFß signaling; (2) induced neural tissue initially acquires an anterior identity (i.e., anterior forebrain); (3) maintenance and expansion of the anterior forebrain depends on the antagonism of posteriorizing signals that would otherwise transform this tissue into posterior neural fates. In this review, we present a historical perspective examining some of the significant experiments that have helped to delineate this molecular model. In addition, we discuss the function of the relevant tissues that act prior to and during gastrulation to ensure proper anterior forebrain formation. Finally, we elaborate data, mainly obtained from the analyses of mouse mutants, supporting a role for transcriptional repressors in the regulation of cell competence within the anterior forebrain. The aim of this review is to provide the reader with a general overview of the signals as well as the signaling centers that control the development of the anterior neural plate.


Asunto(s)
Tipificación del Cuerpo , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Prosencéfalo/metabolismo , Animales , Endodermo/embriología , Endodermo/metabolismo , Gástrula/embriología , Gástrula/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Modelos Moleculares , Placa Neural/embriología , Placa Neural/metabolismo , Prosencéfalo/embriología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Vía de Señalización Wnt
4.
Proc Natl Acad Sci U S A ; 108(28): 11482-7, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21636786

RESUMEN

Wingless (Wnt)/ß-catenin signaling plays an essential role during normal development, is a critical regulator of stem cells, and has been associated with cancer in many tissues. Here we demonstrate that genetic expression of a degradation-resistant mutant form of ß-catenin in early Rathke's pouch (RP) progenitors leads to pituitary hyperplasia and severe disruption of the pituitary-specific transcription factor 1-lineage differentiation resulting in extreme growth retardation and hypopituitarism. Mutant mice mostly die perinatally, but those that survive weaning develop lethal pituitary tumors, which closely resemble human adamantinomatous craniopharyngioma, an epithelial tumor associated with mutations in the human ß-catenin gene. The tumorigenic effect of mutant ß-catenin is observed only when expressed in undifferentiated RP progenitors, but tumors do not form when committed or differentiated cells are targeted to express this protein. Analysis of affected pituitaries indicates that expression of mutant ß-catenin leads to a significant increase in the total numbers of pituitary progenitor/stem cells as well as in their proliferation potential. Our findings provide insights into the role of the Wnt pathway in normal pituitary development and demonstrate a causative role for mutated ß-catenin in an undifferentiated RP progenitor in the genesis of murine and human craniopharyngioma.


Asunto(s)
Hipófisis/citología , Hipófisis/metabolismo , Neoplasias Hipofisarias/etiología , Neoplasias Hipofisarias/metabolismo , Células Madre/citología , Células Madre/metabolismo , Proteínas Wnt/metabolismo , Animales , Diferenciación Celular , Craneofaringioma/etiología , Craneofaringioma/genética , Craneofaringioma/metabolismo , Craneofaringioma/patología , Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Ratones Mutantes , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Hipófisis/crecimiento & desarrollo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , beta Catenina/genética , beta Catenina/metabolismo
5.
PLoS One ; 18(2): e0280001, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36800350

RESUMEN

The chemokine SDF-1 (CXCL12) and its receptor CXCR4 control several processes during embryonic development such as the regulation of stem cell proliferation, differentiation, and migration. However, the role of this pathway in the formation of the pituitary gland is not understood. We sought to characterise the expression patterns of CXCR4, SDF-1 and CXCR7 at different stages of pituitary gland development. Our expression profiling revealed that SDF-1 is expressed in progenitor-rich regions of the pituitary anterior lobe, that CXCR4 and CXCR7 have opposite expression domains and that CXCR4 expression is conserved between mice and human embryos. We then assessed the importance of this signalling pathway in the development and function of the murine pituitary gland through conditional deletion of CXCR4 in embryonic pituitary progenitors. Successful and specific ablation of CXCR4 expression in embryonic pituitary progenitors did not lead to observable embryonic nor postnatal defects but allowed the identification of stromal CXCR4+ cells not derived from HESX1+ progenitors. Further analysis of constitutive SDF-1, CXCR7 and CXCR4 mutants of the pathway indicates that CXCR4 expression in HESX1+ cells and their descendants is not essential for normal pituitary development in mice.


Asunto(s)
Receptores CXCR , Animales , Femenino , Humanos , Ratones , Embarazo , Diferenciación Celular , Quimiocina CXCL12/metabolismo , Embrión de Mamíferos/metabolismo , Proteínas de Homeodominio/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal
6.
Elife ; 82019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30912742

RESUMEN

SOX2 positive pituitary stem cells (PSCs) are specified embryonically and persist throughout life, giving rise to all pituitary endocrine lineages. We have previously shown the activation of the STK/LATS/YAP/TAZ signalling cascade in the developing and postnatal mammalian pituitary. Here, we investigate the function of this pathway during pituitary development and in the regulation of the SOX2 cell compartment. Through loss- and gain-of-function genetic approaches, we reveal that restricting YAP/TAZ activation during development is essential for normal organ size and specification from SOX2+ PSCs. Postnatal deletion of LATS kinases and subsequent upregulation of YAP/TAZ leads to uncontrolled clonal expansion of the SOX2+ PSCs and disruption of their differentiation, causing the formation of non-secreting, aggressive pituitary tumours. In contrast, sustained expression of YAP alone results in expansion of SOX2+ PSCs capable of differentiation and devoid of tumourigenic potential. Our findings identify the LATS/YAP/TAZ signalling cascade as an essential component of PSC regulation in normal pituitary physiology and tumourigenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Hipófisis/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Células Madre/fisiología , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Eliminación de Gen , Redes Reguladoras de Genes , Ratones , Hipófisis/embriología , Hipófisis/crecimiento & desarrollo , Factores de Transcripción SOXB1/análisis , Células Madre/química , Proteínas Señalizadoras YAP
7.
Cell Stem Cell ; 13(4): 433-45, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-24094324

RESUMEN

Sox2(+) adult mouse pituitary cells can self-renew and terminally differentiate in vitro, but their physiological role in vivo and possible contribution to oncogenesis remain largely unknown. Using genetic lineage tracing, we show here that the Sox2(+) cell compartment of both the embryonic and adult pituitary contains stem/progenitor cells that are able to differentiate into all hormone-producing lineages and contribute to organ homeostasis during postnatal life. In addition, we show that targeted expression of oncogenic ß-catenin in Sox2(+) cells gives rise to pituitary tumors, but, unexpectedly, the tumor mass is not derived from the Sox2(+) mutation-sustaining cells, suggesting a paracrine role of Sox2(+) cells in pituitary oncogenesis. Our data therefore provide in vivo evidence of a role for Sox2(+) stem/progenitor cells in long-term physiological maintenance of the adult pituitary, and highlight an unexpected non-cell-autonomous role for these cells in the induction of pituitary tumors.


Asunto(s)
Homeostasis , Hipófisis/citología , Neoplasias Hipofisarias/patología , Factores de Transcripción SOXB1/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Transgénicos , Mutación , Hipófisis/metabolismo , Neoplasias Hipofisarias/metabolismo , Factores de Transcripción SOXB1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA