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











Base de dados
Intervalo de ano de publicação
1.
J Cell Physiol ; 233(2): 1402-1413, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28542730

RESUMO

In this study, we focused on ERß regulation in the adenohypophysis under different estrogenic milieu, by analyzing whether ER modulates the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression and its subcellular localization on anterior pituitary glands from Wistar rats and GH3 lactosomatotroph cells that over-expressed ERß. ERß was regulated in a cyclic manner, and underwent dynamic changes throughout the estrous cycle, with decreased ERß+ cells in estrus and under E2 treatment, but increased in ovariectomized rats. In addition, the ERα/ß ratio increased in estrus and under E2 stimulation, but decreased in ovariectomized rats. Double immunofluorescence revealed that lactotroph and somatotroph ERß+ were significantly decreased in estrus. Also, variations in the PTEN expression was observed, which was diminished with high E2 conditions but augmented with low E2 milieu. The subcellular localization of this phosphatase was cell cycle-dependent, with remarkable changes in the immunostaining pattern: nuclear in arrested pituitary cells but cytoplasmic in stimulated cells, and responding differently to ER agonists, with only DPN being able to increase PTEN expression and retaining it in the nucleus. Finally, ERß over-expression increased PTEN with a noticeable subcellular redistribution, and with a significant nuclear signal increase in correlation with an increase of cells in G0/G1 phase. These results showed that E2 is able to inhibit ERß expression and suggests that the tumoral suppressor PTEN might be one of the signaling proteins by which E2, through ERß, acts to modulate pituitary cell proliferation, thereby adapting endocrine populations in relation with hormonal necessities.


Assuntos
Proliferação de Células , Receptor beta de Estrogênio/metabolismo , Ciclo Estral/metabolismo , Lactotrofos/enzimologia , PTEN Fosfo-Hidrolase/metabolismo , Somatotrofos/enzimologia , Animais , Células Cultivadas , Estradiol/metabolismo , Estradiol/farmacologia , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/genética , Terapia de Reposição de Estrogênios , Feminino , Fase G1 , Lactotrofos/efeitos dos fármacos , Masculino , Nitrilas/farmacologia , Ovariectomia , Ratos Wistar , Transdução de Sinais , Somatotrofos/efeitos dos fármacos , Transfecção
2.
Mol Cell Biochem ; 391(1-2): 67-75, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24553818

RESUMO

This study investigated the roles of Ras, ERK, and Akt in the glucocorticoid-induced differentiation of growth hormone-producing pituitary cells in vitro. Pituicytes isolated from day-18 rat embryos were cultured with 50 mM dexamethasone in addition to specific inhibitors of Ras (manumycin; 0.5, 5, 50 nM), ERK (U0126, 10 µM), or Akt (LY294002, 25 µM). Differentiation was assessed using immunofluorescent staining of intracellular growth hormone. Radioimmunoassay and Western blot analyses were used to determine levels of secreted and intracellular growth hormone, respectively. Manumycin reduced the fraction of growth hormone-positive cells and dexamethasone-induced growth hormone secretion in a dose-dependent manner (both P < 0.001). In the absence of dexamethasone, LY294002 and U0126 did not alter the fraction of growth hormone-positive cells or intracellular growth hormone protein expression or secretion. Both LY294002 and U0126 alone significantly attenuated the fraction of dexamethasone-treated GH-positive cells and the secretion of GH compared to those of cells treated only with dexamethasone (50 nM for 44 h or 48 h) (all P < 0.05). Dexamethasone treatment alone did not change GH protein levels. Treatment of cells with a combination of LY294402 and U0126 significantly attenuated the fraction of dexamethasone-treated GH-positive cells, GH protein levels, and GH secretion compared to cells treated with dexamethasone alone (all P < 0.05). Moreover, dexamethasone-induced phosphorylation of GTP-Ras, ERK, and Akt was significantly attenuated by exposure to the respective inhibitors (P < 0.05). Taken together, our results indicate that Ras, ERK, and Akt are key effectors in the glucocorticoid-induced differentiation of growth hormone-secreting cells.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Embrião de Mamíferos/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glucocorticoides/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Somatotrofos/citologia , Proteínas ras/metabolismo , Animais , Butadienos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Cromonas/farmacologia , Dexametasona/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Hormônio do Crescimento/metabolismo , Morfolinas/farmacologia , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Polienos/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Ratos Sprague-Dawley , Somatotrofos/efeitos dos fármacos , Somatotrofos/enzimologia
3.
Gen Comp Endocrinol ; 166(2): 296-306, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19835876

RESUMO

Previous results suggest that gonadotropin-releasing hormone (GnRH) stimulation of somatotropin secretion in goldfish involves activation of Na(+)/H(+) exchange (NHE). We tested the hypothesis that GnRH alkalinizes intracellular pH (pH(i)) via protein kinase C (PKC) activation of NHE. Two types of alkalinization responses were observed in identified goldfish somatotropes preloaded with the pH-sensitive dye BCECF; the rate of pH(i) changes went from a neutral or slightly negative slope to either a positive or a less negative slope relative to control. Two GnRHs, the PKC-activating TPA, and dioctanoyl glycerol each caused an alkalinization in 70-90% of somatotropes. The PKC inhibitors, Bis II and Gö6976, the NHE inhibitor amiloride, or Na(+)-free solution attenuated TPA and GnRHs actions, suggesting that PKC mediates GnRH activation of NHE. Since amiloride and Na(+)-free solution caused acidification in somatotropes at rest, regulation of basal pH(i) in these cells likely involves Na(+) flux through amiloride-sensitive NHE.


Assuntos
Carpa Dourada/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Proteína Quinase C/fisiologia , Trocadores de Sódio-Hidrogênio/metabolismo , Somatotrofos/metabolismo , Amilorida/farmacologia , Animais , Ativação Enzimática/efeitos dos fármacos , Feminino , Hormônio Liberador de Gonadotropina/administração & dosagem , Concentração de Íons de Hidrogênio , Masculino , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Sódio/metabolismo , Trocadores de Sódio-Hidrogênio/efeitos dos fármacos , Somatotrofos/enzimologia , Acetato de Tetradecanoilforbol/farmacologia
4.
Biochem Biophys Res Commun ; 390(4): 1377-81, 2009 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-19895783

RESUMO

Obestatin, the ghrelin-associated peptide, showed to activate MAPK signaling with no effect on Akt nor cell proliferating activity in rat tumor somatotroph cells (growth cells, GC). A sequential analysis of the obestatin transmembrane signaling pathway indicated a route involving the consecutive activation of G(i), PI3k, novel PKCepsilon, and Src for ERK1/2 activation. Furthermore, obestatin treatment triggers growth hormone (GH) release in the first 30min, being more acute at 15min. At 1h, obestatin treated cells showed the same levels in GH secretion than controls. Added to this functionality, obestatin was secreted by GC cells. Based on the capacity to stimulate GH release from somatotroph cells, obestatin may act directly in the pituitary through an autocrine/paracrine mechanism.


Assuntos
Grelina/farmacologia , Hormônio do Crescimento/metabolismo , Somatotrofos/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Ativação Enzimática , Camundongos , Proteína Quinase 3 Ativada por Mitógeno/biossíntese , Fosfatidilinositol 3-Quinases/biossíntese , Proteína Quinase C-épsilon/biossíntese , Ratos , Somatotrofos/enzimologia , Somatotrofos/metabolismo , Quinases da Família src/biossíntese
5.
Mol Cell Endocrinol ; 296(1-2): 78-86, 2008 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-18824214

RESUMO

We investigated the pattern of extracellular signal-regulated kinase (ERK) phosphorylation and the induction of mitogen-activated protein kinase phosphatase 1 (MKP-1) by thyrotropin-releasing hormone (TRH) under various stimulation conditions in pituitary GH3 cells. In static culture, ERK activation by continuous TRH was maximal at 10 min and persisted for up to 60 min, with a return to the basal level by 2h. Stimulation with continuous TRH in perifused cells resulted in a similar level of ERK phosphorylation. MKP-1 was expressed 60 min following either static or perifused, continuous TRH stimulation. When cells were stimulated with pulsatile TRH every 30 min, ERK activation was maximal at 10 min and returned to its baseline level by 30 min. ERK was phosphorylated again with each subsequent pulse. Pulsatile TRH did not induce MKP-1. Prolactin promoter activity following continuous, static TRH stimulation was higher than that following perifused TRH stimulation. TRH at a frequency of one pulse every 30 min increased prolactin promoter activity similar to that of perifused, continuous TRH stimulation. Additionally, changes in pulse frequency resulted in alterations in the level of prolactin promoter. Following static stimulation, a 10 min exposure to TRH was sufficient to obtain full activation of the prolactin promoter. Additionally, a 5-10 min exposure of TRH was sufficient to maintain ERK activation. A single 5-min pulse of TRH stimulation resulted in low activation of the prolactin promoter. ERK activation was necessary for prolactin gene transcription; however, prolactin gene transcription is not entirely determined by the strength or duration of TRH-induced ERK activation.


Assuntos
Fosfatase 1 de Especificidade Dupla/biossíntese , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Somatotrofos/efeitos dos fármacos , Hormônio Liberador de Tireotropina/farmacologia , Algoritmos , Animais , Butadienos/farmacologia , Técnicas de Cultura de Células , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , Nitrilas/farmacologia , Prolactina/genética , Prolactina/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Fluxo Pulsátil/fisiologia , Ratos , Somatotrofos/enzimologia , Somatotrofos/metabolismo , Hormônio Liberador de Tireotropina/administração & dosagem
6.
J Cell Physiol ; 215(1): 140-50, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17941086

RESUMO

To investigate the mechanisms by which the hypothalamic peptide GHRH influences cell division, we analyzed its effects on the proliferation of two different cell lines: CHO-4, an ovary-derived cell line, and GH3, a pituitary-derived cell line. We found that GHRH induces the proliferation of pituitary-derived cells but inhibits the proliferation of ovary-derived cells. We further characterized this dual effect of GHRH to find that the cytoplasmic signals induced by this hormone are similar in both cell lines. Moreover, in CHO-4 cells GHRH stimulates two well-known positive cell cycle regulators, c-myc and cyclin D1, but is unable to induce the degradation of the negative cell cycle regulator p27(Kip1). Significantly, when the Pit-1/GHF-1 gene is exogenously expressed in CHO-4 cells, the negative effect of GHRH on the proliferation of these cells is attenuated. Furthermore, when the levels of Pit-1 are downregulated by siRNA in GH3-GHRHR cells, the positive effects of GHRH on the proliferation of these cells are diminished. These findings add to our understanding of the molecules involved in the regulation of cell proliferation by GHRH, as we demonstrate for the first time that Pit-1 is not only required to drive the expression of the GHRH receptor, as previously described, but is also needed for the downstream effects that occur after its activation to modulate cell proliferation. These data suggest that the regulation of cell proliferation in response to a specific growth factor depends in certain cell populations on the presence of a tissue-specific transcription factor.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/farmacologia , Somatotrofos/citologia , Somatotrofos/efeitos dos fármacos , Fator de Transcrição Pit-1/metabolismo , Animais , Células CHO , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cricetinae , Cricetulus , AMP Cíclico/biossíntese , Ciclina D1/genética , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação para Baixo/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Especificidade de Órgãos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/genética , Ratos , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Fase S/efeitos dos fármacos , Soro , Somatotrofos/enzimologia , Transcrição Gênica/efeitos dos fármacos
7.
Endocrinology ; 148(6): 2973-83, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17363453

RESUMO

In pituitary cells, activation of the cAMP pathway by specific G protein-coupled receptors controls differentiative functions and proliferation. Constitutively active forms of the alpha subunit of the heterotrimeric G(s) protein resulting from mutations at codon 201 or 227 (gsp oncogene) were first identified in 30-40% of human GH-secreting pituitary adenomas. This rate of occurrence suggests that the gsp oncogene is not responsible for initiating the majority of these tumors. Moreover, there is a large overlap between the clinical phenotypes observed in patients with tumors bearing the gsp oncogene and those devoid of this oncogene. To explore the role of G(s)alpha in GH-secreting adenomas, we obtained somatolactotroph GH4C1 cell lines by performing doxycycline-dependent conditional overexpression of the wild-type G(s)alpha protein and expression of the gsp oncogene. Although the resulting adenylyl cyclase and cAMP levels were 10-fold lower in the wild-type G(s)alpha-overexpressing cell line, a sustained MAPK ERK1/2 activation was observed in both cell lines. Overexpression of the wild-type G(s)alpha protein as the gsp oncogene initiated chronic activation of endogenous prolactin synthesis and release, as well as chronic activation of ERK1/2-sensitive human prolactin and GH promoters.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Regulação Enzimológica da Expressão Gênica , Hormônio do Crescimento/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Prolactina/metabolismo , Somatotrofos/metabolismo , Adenilil Ciclases/metabolismo , Animais , Linhagem Celular , Cromograninas , AMP Cíclico/metabolismo , Doxiciclina/farmacologia , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hormônio do Crescimento/genética , Proteínas Oncogênicas/genética , Prolactina/genética , Ratos , Somatotrofos/enzimologia , Fatores de Tempo , Transfecção , Transgenes/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA