Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Endocrinology ; 159(6): 2421-2434, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29726995

RESUMEN

Prolactin-secreting adenomas, or prolactinomas, cause hypogonadism, osteoporosis, and infertility. Although dopamine agonists (DAs) are used clinically to treat prolactinoma and reduce prolactin secretion via cAMP inhibition, the precise mechanism by which DAs inhibit lactotrope proliferation has not been defined. In this study, we report that phosphatidylinositol 3-kinase (PI3K) signals through AKT and mTOR to drive proliferation of pituitary somatolactotrope GH4T2 cells. We demonstrate that the DA cabergoline reduces activity of the mTOR effector s6K and diminishes GH4T2 cell proliferation primarily via activation of the long isoform of the dopamine D2 receptor (D2R). Dysfunctional D2R-mediated signaling and/or downregulated D2R expression is thought be the primary mechanism of DA resistance, which is observed in 10% to 20% of prolactinoma tumors. Dopamine-mediated D2R activation results in ERK stimulation and PI3K inhibition, suggesting that these two pathways act in an inverse manner to maintain lactotrope homeostasis. In this study, we found that ERK1/2-mediated prolactin transcription is inhibited by PI3K/CDK4-driven cell cycle progression, emphasizing that the ERK and PI3K signaling pathways oppose one another in lactotrope cells under homeostatic conditions. Lastly, we show that both ERK1/2 and AKT are activated in prolactinoma, demonstrating that the balance of ERK and AKT is dysregulated in human prolactinoma. Our findings reveal a potential use for dual pharmacological inhibitors of ERK and AKT as an alternative treatment strategy for DA-resistant prolactinomas.


Asunto(s)
Dopamina/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasa/metabolismo , Neoplasias Hipofisarias/metabolismo , Prolactinoma/metabolismo , Animales , Cabergolina/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Células HEK293 , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/genética , Fosfatidilinositol 3-Quinasa/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Prolactinoma/genética , Prolactinoma/patología , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
2.
Best Pract Res Clin Endocrinol Metab ; 32(3): 219-239, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29779578

RESUMEN

Gonadotropins are pituitary gonadotrope-derived glycoprotein hormones. They act by binding to G-protein coupled receptors on gonads. Gonadotropins play critical roles in reproduction by regulating both gametogenesis and steroidogenesis. Although biochemical and physiological studies provided a wealth of knowledge, gene manipulation techniques using novel mouse models gave new insights into gonadotropin synthesis, secretion and action. Both gain of function and loss of function mouse models for understanding gonadotropin action in a whole animal context have already been generated. Moreover, recent studies on gonadotropin actions in non-gonadal tissues challenged the central dogma of classical gonadotropin actions in gonads and revealed new signaling pathways in these non-gonadal tissues. In this Chapter, we have discussed our current understanding of gonadotropin synthesis, secretion and action using a variety of genetically engineered mouse models.


Asunto(s)
Gonadotropinas/metabolismo , Gonadotropinas/fisiología , Ratones , Modelos Animales , Animales , Hormona Folículo Estimulante , Humanos , Reproducción/fisiología
3.
Mol Cell Endocrinol ; 476: 165-172, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29753028

RESUMEN

Distinct cell types have been shown to respond to activated Ras signaling in a cell-specific manner. In contrast to its pro-tumorigenic role in some human epithelial cancers, oncogenic Ras triggers differentiation of pheochromocytoma cells and medullary thyroid carcinoma cells. Furthermore, we have previously demonstrated that in pituitary somatolactotropes, activated Ras promotes differentiation and is not sufficient to drive tumorigenesis. These findings demonstrate that lactotrope cells have the ability to evade the tumorigenic fate that is often associated with persistent activation of Ras/ERK signaling, and suggest that there may be differential expression of inhibitory signaling molecules or negative cell cycle regulators that act as a brake to prevent the tumorigenic effects of sustained Ras signaling. Here we aim to gain further insight into the mechanisms that allow GH4T2 cells to evade an oncogenic response to Ras. We show that Ral, but likely not menin, plays a key role in directing Ras-mediated differentiation of somatolactotropes, which may allow these cells to escape the tumorigenic fate that is often associated with activated Ras signaling. We also show that dominant negative Ras expression results in reduced GH4T2 cell proliferation and transformation, but does not influence differentiation. Taken together, the data presented here begin to shed light on the mechanisms by which pituitary somatolactotropes evade an oncogenic response to persistently activated Ras signaling and suggest that the architecture of the Ras signaling cascade in some endocrine cell types may be distinct from that of cells that respond to Ras in an oncogenic manner.


Asunto(s)
Carcinogénesis/metabolismo , Carcinogénesis/patología , Lactotrofos/metabolismo , Oncogenes , Hipófisis/metabolismo , Proteínas ras/metabolismo , Animales , Proliferación Celular , Prolactina/genética , Prolactina/metabolismo , Regiones Promotoras Genéticas/genética , Ratas , Factores de Transcripción/metabolismo
4.
Mol Cell Endocrinol ; 463: 87-96, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-28445712

RESUMEN

Conserved signaling pathways are critical regulators of pituitary homeostasis and, when dysregulated, contribute to adenoma formation. Pituitary adenomas are typically benign and rarely progress to malignant cancer. Pituitary and other neuroendocrine cell types often display non-proliferative responses to ERK and PI3K, in contrast to non-endocrine cell types which typically proliferate in response to ERK and PI3K activation. These differences likely contribute to the infrequent progression to malignancy in many endocrine tumors. In this review, we highlight the Ras/ERK and PI3K/AKT/mTOR signaling pathways in each pituitary cell type, as well as in other endocrine tissues. Furthermore, we provide evidence that a balance of ERK and PI3K signaling is required to maintain pituitary homeostasis. It is unlikely that one sole oncogene will be identified as being responsible for sporadic pituitary adenoma formation. This review emphasizes the necessity to consider endocrine cell-specific contexts and the interplay of signaling pathways to define the mechanisms underlying pituitary tumorigenesis.


Asunto(s)
Hipófisis/metabolismo , Hipófisis/patología , Transducción de Señal , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Humanos , Modelos Biológicos , Sistemas Neurosecretores/metabolismo , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA