Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Clin Invest ; 87(1): 326-30, 1991 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1985104

RESUMO

To test the hypothesis that uterine decidua may modulate trophoblast function, trophoblasts and decidual cells were isolated from term placentas by enzymatic digestion and Percoll gradient centrifugation. Placental trophoblasts were cocultured with decidual cells and trophoblasts or JEG-3 choriocarcinoma cells were incubated with medium conditioned by decidual cells (DCM) for 72-96 h. In cocultures decidual cells inhibited choriogonadotropin (hCG) release from trophoblasts by 75% in comparison with controls (P less than 0.001). The DCM contained a factor that markedly inhibited hCG release from trophoblasts and JEG cells in vitro compared with controls. The inhibitory effect of the factor on hCG release was dose dependent, and could be eliminated by boiling the DCM for 30 min or proteolytic enzyme treatment. Ultrafiltration and Sephadex G-50 fractionation of the DCM indicated that the apparent molecular mass was 7,000-10,000 D. DCM also inhibited the stimulatory effect of exogenous cAMP on hCG secretion by JEG-3 cells, suggesting that DCM may interfere with activation of the cAMP-dependent protein kinases or transcription of hCG genes. These results suggest that the release of trophoblast hCG is under local paracrine control, regulated in part by a protein released by decidual cells.


Assuntos
Gonadotropina Coriônica/metabolismo , Decídua/metabolismo , Proteínas/fisiologia , Trofoblastos/metabolismo , Células Cultivadas , Feminino , Humanos , Peso Molecular , Gravidez
2.
J Clin Invest ; 97(8): 1852-9, 1996 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-8621768

RESUMO

We recently described the expression of leukemia inhibitory factor (LIF) in human fetal and murine corticotrophs. LIF and the related cytokine oncostatin M induced basal, and corticotropin-releasing hormone (CRH) induced proopiomelanocortin (POMC) mRNA and ACTH secretion in AtT20 cells. LIF signaling and regulation of POMC gene transcription were therefore tested. Dexamethasone inhibited both basal- and LIF-induced ACTH secretion (P<0.05) and LIF induction of ACTH was also attenuated by immuneutralization of either the LIF receptor (35%, P<0.05) or the gp130 affinity converter (41%, P<0.05). These antisera also attenuated basal ACTH secretion in the absence of added ligand (P<0.05). To examine intrapituitary LIF signaling, phosphorylation of post-receptor substrates was measured. 1 nM LIF rapidly induced tyrosyl phosphorylation of STAT 1 and STAT 3 proteins, as well as tyrosyl phosphorylation of a 115-kD protein, coimmunoprecipitated with STAT 1. The transfected rat POMC promoter -706/+64, fused to the luciferase reporter gene, was induced by LIF, which exerted strong (18-fold) synergy with CRH. Deletion of the major CRH responsive region in POMC (-323/-166) abolished CRH induction of transcription and severely limited LIF synergy. Although 8 bromo cAMP or forskolin modestly enhanced POMC transcription (2.8-fold), LIF markedly potentiated (7.4-fold) these cAMP activators. These results demonstrate that corticotroph LIF action is receptor mediated and involves activation of STAT signaling pathways. LIF potently synergizes with both CRH and cAMP induction of POMC transcription. This novel intrapituitary signaling mechanism may mediate a neuroimmune pituitary interface.


Assuntos
Expressão Gênica/efeitos dos fármacos , Inibidores do Crescimento/farmacologia , Linfocinas/farmacologia , Pró-Opiomelanocortina/biossíntese , Transdução de Sinais , Ativação Transcricional , Hormônio Adrenocorticotrópico/biossíntese , Hormônio Adrenocorticotrópico/metabolismo , Animais , Linhagem Celular , Hormônio Liberador da Corticotropina/farmacologia , Dexametasona/farmacologia , Feto , Humanos , Interleucina-6/farmacologia , Cinética , Fator Inibidor de Leucemia , Subunidade alfa de Receptor de Fator Inibidor de Leucemia , Luciferases/biossíntese , Oncostatina M , Peptídeos/farmacologia , Hipófise/metabolismo , Neoplasias Hipofisárias , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Receptores de Citocinas/imunologia , Receptores de Citocinas/fisiologia , Receptores de OSM-LIF , Proteínas Recombinantes de Fusão/biossíntese , Transfecção , Células Tumorais Cultivadas
3.
J Clin Invest ; 95(3): 1288-98, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7883977

RESUMO

Leukemia inhibitory factor (LIF) gene expression was detected in human fetal pituitary tissue by expression of LIF mRNA transcripts, protein immunocytochemistry, and immunoelectron microscopy. Fetal LIF immunoreactivity colocalized with 30% of ACTH-expressing cells, approximately 20% of somatotrophs, and approximately 15% of non-hormone-expressing cells. LIF was also strongly expressed in normal adult pituitary and in four growth hormone-producing and two ACTH-producing adenomas, but not in eight nonfunctioning pituitary tumors. Culture of fetal cells expressing surface LIF-binding sites demonstrated predominance of in vitro ACTH secretion as compared with other pituitary hormones. In AtT-20 murine cells, LIF (ED50 10 pM) stimulated basal proopiomelanocortin mRNA levels by 40% and corticotropin-releasing hormone-induced ACTH secretion (two- to threefold), as did oncostatin M (ED50 30 pM), a related peptide. ACTH responses were not further enhanced by both cytokines together, which is consistent with their shared receptor. Anti-LIF antiserum neutralized basal and LIF-induced ACTH secretion, suggesting autocrine regulation of ACTH by LIF. The results show that human pituitary cells express the LIF gene and LIF-binding sites, predominantly in corticotrophs. Pituitary LIF expression and LIF regulation of proopiomelanocortin and ACTH reflect an intrapituitary role for LIF in modulating early embryonic determination of specific human pituitary cells and as a paracrine or autocrine regulator of mature ACTH.


Assuntos
Inibidores do Crescimento/biossíntese , Interleucina-6 , Linfocinas/biossíntese , Hipófise/metabolismo , Adenoma/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Animais , Células Cultivadas , Hormônio Liberador da Corticotropina/farmacologia , Relação Dose-Resposta a Droga , Citometria de Fluxo , Inibidores do Crescimento/genética , Inibidores do Crescimento/imunologia , Inibidores do Crescimento/farmacologia , Humanos , Imuno-Histoquímica , Fator Inibidor de Leucemia , Linfocinas/genética , Linfocinas/imunologia , Camundongos , Microscopia Imunoeletrônica , Testes de Neutralização , Oncostatina M , Peptídeos/farmacologia , Hipófise/efeitos dos fármacos , Hipófise/embriologia , Hipófise/ultraestrutura , Neoplasias Hipofisárias/metabolismo , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/genética , RNA Mensageiro/análise
4.
J Endocrinol ; 189(3): 519-28, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16731783

RESUMO

Male mice that are pttg-null develop sexually dimorphic diabetes with hypoinsulinemia secondary to reduced postnatal-cell proliferation and an inability to expand islet cell mass with aging. We therefore examined the effects of sex-steroid manipulation on diabetes development in pttg-/- male mice. Surgical gonadectomy was followed by implantation of 90-day slow-release pellets releasing 17beta-estradiol (0.36 mg/pellet), placebo or dihydrotestosterone (DHT; 12.5 mg/pellet). Mean fasting blood sugars at the end of the study were 414 +/- 54 mg/dl for pttg-/- controls and 371 +/- 14 mg/dl for pttg-/- mice gonadectomized and treated with DHT compared with 124 +/- 40 and 85 +/- 12 mg/dl in gonadectomized pttg-/- males treated with placebo or estradiol, respectively (P < 0.01 compared with control pttg-/-). Gonadectomy with and without estradiol treatment did not increase the very low circulating insulin levels in pttg-null males (fasting insulin 0.44 +/- 0.04 ng/ml in pttg-/- controls, 0.47 +/- 0.07 and 0.4 ng/ml in pttg-/- gonadectomized males treated with placebo or estradiol, respectively). Gonadectomy increased serum adiponectin levels (4.9 +/- 008 microg/ml in pttg-/- controls versus 13 +/- 0.08 and 7.5 +/- 0.6 microg/ml in pttg-/- gonadectomized males treated with placebo or estradiol, respectively; P < 0.001 and P < 0.05), accompanied by increased insulin sensitivity. The results show that gonadectomy delayed, and gonadectomy with additional estradiol treatment prevented, diabetes development in pttg-/- males, possibly through increased insulin sensitivity mediated by elevated serum adiponectin levels. Male-selective effects of disrupted beta-cell proliferation in the absence of pttg are restored by sex-steroid effects on peripheral insulin sensitivity.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Hormônios Esteroides Gonadais/uso terapêutico , Proteínas de Neoplasias/metabolismo , Adiponectina/sangue , Androgênios/uso terapêutico , Animais , Glicemia/análise , Peptídeo C/sangue , Proteínas de Transporte/metabolismo , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Di-Hidrotestosterona/uso terapêutico , Estradiol/uso terapêutico , Insulina/sangue , Células Secretoras de Insulina/patologia , Leptina/sangue , Masculino , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Orquiectomia , Securina , Caracteres Sexuais
5.
Mol Endocrinol ; 12(11): 1708-20, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9817597

RESUMO

Leukemia inhibitory factor (LIF) regulates the mature hypothalamic-pituitary-adrenal axis in vivo. In vitro, LIF determines corticotroph cell proliferation and induces POMC transcription. To explore LIF action on pituitary development, transgenic mice expressing LIF driven by the pituitary glycoprotein hormone alpha-subunit (alphaGSU) promoter were generated. Transgenic mice exhibited dwarfism with low IGF-I (29 +/- 9 ng/ml vs. wild type (WT) 137 +/- 16 ng/ml; P < 0.001), hypogonadism with low FSH (0.04 +/- 0.023 ng/ml vs. WT 0.63 +/- 0.18 ng/ml; P < 0.001), and Cushingoid features of thin skin and truncal obesity with elevated cortisol levels (86 +/- 22 ng/ml vs. WT 50 +/- 14 ng/ml; P = 0.002). Their pituitary glands showed corticotroph hyperplasia, striking somatotroph and gonadotroph hypoplasia, and multiple Rathke-like cysts lined by ciliated cells. LIF, overexpressed in Rathke's pouch at embryonal day 10, diverts the differentiation stream of hormone-secreting cells toward the corticotroph lineage and ciliated nasopharyngeal-like epithelium. Thus, inappropriate expression of LIF, a neuro-immune interfacing cytokine, plays a key role in the terminal differentiation events of pituitary development and mature pituitary function.


Assuntos
Síndrome de Cushing/etiologia , Inibidores do Crescimento/toxicidade , Interleucina-6 , Linfocinas/toxicidade , Neuroimunomodulação , Hipófise/embriologia , Sistema Hipófise-Suprarrenal/fisiopatologia , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Síndrome de Cushing/genética , Dexametasona , Nanismo Hipofisário/etiologia , Nanismo Hipofisário/genética , Feminino , Hormônio Foliculoestimulante/deficiência , Inibidores do Crescimento/biossíntese , Inibidores do Crescimento/genética , Inibidores do Crescimento/farmacologia , Hiperplasia , Hipogonadismo/etiologia , Hipogonadismo/genética , Infertilidade/etiologia , Infertilidade/genética , Fator de Crescimento Insulin-Like I/deficiência , Fator Inibidor de Leucemia , Linfocinas/biossíntese , Linfocinas/genética , Linfocinas/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Hipófise/crescimento & desenvolvimento , Hipófise/metabolismo , Hipófise/patologia , Pró-Opiomelanocortina/biossíntese , Pró-Opiomelanocortina/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/toxicidade , Células-Tronco/metabolismo , Células-Tronco/patologia , Transgenes
6.
Mol Endocrinol ; 14(8): 1137-46, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10935539

RESUMO

The pituitary transforming gene, PTTG, is abundantly expressed in endocrine neoplasms. PTTG has recently been recognized as a mammalian securin based on its biochemical homology to Pds1p. PTTG expression and intracellular localization were therefore studied during the cell cycle in human placental JEG-3 cells. PTTG mRNA and protein expressions were low at the G1/S border, gradually increased during S phase, and peaked at G2/M, but PTTG levels were attenuated as cells entered G1. In interphase cells, wild-type PTTG, an epitope-tagged PTTG, and a PTTG-EGFP conjugate all localized to both the nucleus and cytoplasm, but in mitotic cells, PTTG was not observed in the chromosome region. PTTG-EGFP colocalized with mitotic spindles in early mitosis and was degraded in anaphase. Intracellular fates of PTTG-EGFP and a conjugate of EGFP and a mutant inactivated PTTG devoid of an SH3-binding domain were observed by real-time visualization of the EGFP conjugates in live cells. The same cells were continuously observed as they progressed from G1/S border to S, G2/M, and G1. Most cells (67%) expressing PTTG-EGFP died by apoptosis, and few cells (4%) expressing PTTG-EGFP divided, whereas those expressing mutant PTTG-EGFP divided. PTTG-EGFP, as well as the mutant PTTG-EGFP, disappeared after cells divided. The results show that PTTG expression and localization are cell cycle-dependent and demonstrate that PTTG regulates endocrine tumor cell division and survival.


Assuntos
Ciclo Celular/fisiologia , Proteínas de Neoplasias/metabolismo , Placenta/citologia , Sequência de Aminoácidos , Divisão Celular/fisiologia , Sobrevivência Celular/fisiologia , Proteínas de Fluorescência Verde , Humanos , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Securina , Células Tumorais Cultivadas
7.
Endocrinology ; 128(3): 1623-9, 1991 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1999176

RESUMO

Recent studies have shown that insulin regulates placental lactogen, progesterone, and estrogen production from human trophoblast cells. This study was performed to examine whether insulin also regulates the production of hCG by this type of cell. After 24-36 h of preincubation, JEG-3 and JAR cells (2-3 x 10(5) cells/ml.well) or human term trophoblast cells (1 x 10(6) cells/ml.well) were exposed to the test hormone in serum-free Dulbecco's Modified Eagle's Medium for 24-96 h. Secretion of hCG from JEG-3 cells was stimulated by human insulin, human proinsulin, or porcine insulin in a dose-dependent manner, with lowest effective doses of 6.7, 96, and 53 mg/L, respectively. Time-course studies showed that hCG secretion peaked at 72-96 h with insulin exposure; in contrast, no decernable peak was seen without insulin in serum-free media. Exposure of JEG-3 cells for 24 h to 209 mg/liter insulin stimulated hCG synthesis, with 40 +/- 3% more immunoreactive intracellular hCG (P less than 0.05). Cells grown in the presence of insulin and [35S]methionine had 47 +/- 21% more labeled intracellular hCG and 56 +/- 13% more immunoprecipitable [35S]methionine-hCG secreted into the medium than the control cultures (P less than 0.05). During this time period, human placental lactogen release and total trichloroacetice acid-precipitable [35S]methionine protein were not increased. The insulin-induced stimulation of hCG synthesis was inhibited by cycloheximide. Additionally, insulin did not significantly affect total intracellular protein during 24-96 h of incubation. Insulin also increased hCG release from JAR cells, but not from human term trophoblast cells. A mouse monoclonal antibody to the IGF-I receptor inhibited the stimulation of insulin in JEG-3 cells. We conclude that insulin stimulates the synthesis and secretion of hCG from JEG-3 cells and JAR cells, and that hCG regulation in choriocarcinoma cells differs from that in primary human placental trophoblast cells. The effect of insulin on JEG-3 cells may be mediated in part through the insulin-like growth factor-I receptor.


Assuntos
Coriocarcinoma/metabolismo , Gonadotropina Coriônica/metabolismo , Insulina/farmacologia , Animais , Coriocarcinoma/patologia , Gonadotropina Coriônica/biossíntese , Relação Dose-Resposta a Droga , Humanos , Insulina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Radioisótopos do Iodo , Suínos , Fatores de Tempo , Células Tumorais Cultivadas
8.
Endocrinology ; 137(7): 2947-53, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8770918

RESUMO

We have recently shown expression of leukemia inhibitory factor (LIF) in human fetal pituitary tissue and its in vitro induction of POMC transcription. We now use qualitative and semiquantitative RT-PCR to demonstrate that LIF and LIF-receptor (LIF-R) are constitutively expressed in the normal mouse hypothalamus and pituitary. Hypothalamic and pituitary LIF and LIF-R are significantly induced (up to 6- and 4-fold, respectively) in vivo in response to lipopolysaccharide endotoxin (LPS) administered to B6D2F1 and C57BL/6 mice. In contrast to the nearly exclusive expression of matrix-associated LIF messenger RNA (mRNA) in control hypothalamus and pituitary, both diffusible and matrix-associated LIF mRNA alternate transcripts are induced by LPS. Furthermore, the time course of peripheral ACTH-response to LPS peaks at 60 min, whereas hypothalamic LIF mRNA increase occurs at 30 min and pituitary LIF induction occurs at 60 min. These results show that mLIF is a novel LPS-inducible proinflammatory neuroendocrine cytokine and the alternatively spliced diffusible LIF may play a paracrine role in activating pituitary ACTH secretion in synergy with hypothalamic CRH, implying a mechanism for central nervous system cytokine responses to immune signals.


Assuntos
Inibidores do Crescimento/biossíntese , Hipotálamo/metabolismo , Interleucina-6 , Lipopolissacarídeos/farmacologia , Linfocinas/biossíntese , Hipófise/metabolismo , Transcrição Gênica , Animais , Sequência de Bases , Encéfalo/metabolismo , Primers do DNA , Endotoxinas/farmacologia , Escherichia coli , Humanos , Hipotálamo/efeitos dos fármacos , Fator Inibidor de Leucemia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Dados de Sequência Molecular , Especificidade de Órgãos , Hipófise/efeitos dos fármacos , Reação em Cadeia da Polimerase , Pró-Opiomelanocortina/biossíntese , RNA Mensageiro/biossíntese , Mapeamento por Restrição , Especificidade da Espécie , Transcrição Gênica/efeitos dos fármacos
9.
Endocrinology ; 131(5): 2479-81, 1992 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1385103

RESUMO

As the somatostatin analog octreotide suppresses pituitary GH secretion and circulating IGF-1 levels, we examined its effects on human hepatoma (hep G2) cells which selectively express IGFBP-1. Octreotide (60 nM) stimulated IGFBP-1 up to 4.1-fold (p < 0.001 after 24 hrs). Induction of IGFBP-1 was first detectable after 12 hrs of 6 nM octreotide (1.5-fold, p < 0.03), and was confirmed by ligand blotting. Cholera toxin and forskolin induced IGFBP-1 independently and were also additive with octreotide. IGFBP-1 mRNA expression was induced 2.7-fold by octreotide. Thus, octreotide induces basal and stimulated IGFBP-1 in hepatocytes independently of insulin and GH. As IGFBP-1 may regulate peripheral IGF-1 action, induction of IGFBP-1 represents a novel pituitary-independent mechanism for octreotide action.


Assuntos
Carcinoma Hepatocelular/química , Proteínas de Transporte/análise , Neoplasias Hepáticas/química , Octreotida/farmacologia , Somatostatina/análogos & derivados , Northern Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Toxina da Cólera/farmacologia , Colforsina/farmacologia , Eletroforese em Gel de Poliacrilamida , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , RNA Mensageiro/análise , RNA Mensageiro/genética , Radioimunoensaio , Células Tumorais Cultivadas/química , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia
10.
J Clin Endocrinol Metab ; 70(4): 983-9, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2318953

RESUMO

To investigate the regulation of the synthesis and secretion of placental proteins-12 (PP12) and -14 (PP14) and PRL, explants and enriched preparations of stromal cells and gland cells obtained from 10 human early pregnancy decidua were preincubated in medium for 24 h (baseline), followed by incubation in medium with or without progesterone (0.02-32 mumol/L), hCG (10 and 100 ng/ml), or cAMP (0.25-1 mmol/L) in an atmosphere of 5% CO2-95% air at 37 C for another 96-120 h. Media were changed each 24 h, and PP12, PP14, and PRL levels were determined by RIA. Decidual explants, as well as their isolated cells produced detectable levels of PP12, PP14, and PRL in vitro. The gland cells synthesized and secreted about 30 times more PP14 than did stromal cells. After 96-120 h of incubation, the production of each protein by control cultures was increased 81-167% compared to the baseline (not significant). The secretion of these proteins in medium supplemented with progesterone or hCG was not significantly different from that in the control groups. 8-Bromo-cAMP significantly increased the secretion of PRL and PP12, but not PP14, by stromal cells compared to control values. We conclude that 1) PP14 is mainly produced by decidual gland cells; 2) progesterone at the concentrations used in our study does not stimulate production of PP12, PP14, and PRL in decidualized endometrium in vitro; 3) hCG does not stimulate the production of PP12 and PP14 in decidualized endometrium; and 4) 8-bromo-cAMP stimulates decidual stromal cell secretion of PRL and PP12.


Assuntos
Gonadotropina Coriônica/farmacologia , Decídua/efeitos dos fármacos , Glicoproteínas , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Proteínas da Gravidez/biossíntese , Progesterona/farmacologia , Prolactina/biossíntese , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Adulto , Células Cultivadas , Técnicas de Cultura , Decídua/metabolismo , Relação Dose-Resposta a Droga , Endométrio/metabolismo , Feminino , Glicodelina , Humanos , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina , Radioimunoensaio
11.
J Clin Endocrinol Metab ; 73(2): 441-3, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1713220

RESUMO

Insulin-like growth factors (IGFs) circulate in a complexed state with several binding proteins (BPs). Of these, IGFBP-1 is regulated by hormonal and nutritional factors. The somatostatin analogue, octreotide, has been used to effectively control hypersomaototropism in acromegaly. IGFBP-1 levels were measured by RIA in 17 acromegalic patients receiving octreotide. Serum hormone sampling was conducted hourly for 8 hr periods. Among 13 octreotide responders, mean pre-treatment basal GH, IGF-1, and IGFBP-1 levels were 19 +/- 5 micrograms/L, 1021 +/- 168 micrograms/L, and 36 +/- 8 micrograms/L respectively. One month following octreotide treatment, an acute subcutaneous injection (100 micrograms) maximally attenuated GH to 3 +/- 0.6 microgram/L (18% of control, P less than 0.03) and IGF-1 to 467 +/- 75 micrograms/L (46% of control, P less than 0.008) after 4 hrs. IGFBP-1 levels, however, were stimulated to 95 +/- 16 micrograms/L (297% of control, P less than 0.003) during the same time period. A significant increase in IGFBP-1 levels occurred within 2 hrs (158% of baseline, P less than 0.03), and was sustained until the 7th hr following injection. Insulin, a known suppressor of IGFBP-1, did not change during this time. Among the 4 octreotide non-responders, mean basal IGFBP-1 levels were 42 +/- 4 micrograms/L, and 4 hrs following octreotide administration IGFBP-1 was 40 +/- 7 micrograms/L. Octreotide induced a dynamic inverse relationship between circulating GH and IGFBP-1 levels (r = -0.73, P less than 0.001). The absence of IGFBP-1 changes in octreotide non-responders and the non-suppression of insulin in octreotide responsive patients, suggest a direct GH-mediated mechanism of IGFBP-1 regulation in octreotide treated patients with acromegaly. IGFBP-1 may be another useful marker in evaluating the response of acromegaly to octreotide treatment in patients who experience clinical benefit but equivocal GH and IGF-1 attenuation.


Assuntos
Acromegalia/sangue , Proteínas de Transporte/sangue , Hormônio do Crescimento/sangue , Fator de Crescimento Insulin-Like I/metabolismo , Octreotida/uso terapêutico , Acromegalia/tratamento farmacológico , Proteínas de Transporte/efeitos dos fármacos , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Radioimunoensaio , Fatores de Tempo
12.
J Clin Endocrinol Metab ; 75(6): 1459-63, 1992 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1281485

RESUMO

As the long-acting somatostatin analog octreotide attenuates polypeptide hormone hypersecretion, it has recently been used to effectively treat acromegaly and gastrointestinal carcinoid tumors. Most growth-promoting actions of GH are mediated by insulin-like growth factor-I (IGF-I), which circulates complexed with multiple binding proteins (IGFBPs). IGFBP-1, a nonglycosylated peptide, competes with the IGF-I receptor for ligand binding and also regulates IGF action. To examine GH-independent mechanisms for octreotide regulation of the GH axis, circulating levels of IGFBP-1 were measured hourly after sc octreotide or saline administration in normal and GH-deficient adults. As IGFBP-1 is inhibited by insulin and GH, the dynamic pattern of alterations in GH and insulin levels was also assessed. After octreotide (100 micrograms) administration to 10 normal subjects, mean IGFBP-1 concentrations were stimulated from 23 +/- 4 to 72 +/- 18 micrograms/L (P < 0.007 vs. saline) after 2 h. Maximal induction of IGFBP-1 levels occurred after 3 h (325 +/- 115 micrograms/L; P < 0.02 vs. saline) and remained elevated (P < 0.005) for 6 h. IGFBP-1 was induced by octreotide in all subjects and was confirmed by Western ligand blotting. Insulin and GH levels preceding the rise in IGFBP-1 were unaltered by octreotide. Octreotide stimulated IGFBP-1 5-fold during a sustained fast in 4 normal subjects, despite equally suppressed insulin levels in both saline- and octreotide-treated groups. In 4 GH-deficient adults, IGFBP-1 levels were stimulated by octreotide from 16 +/- 3 to 146 +/- 36 and 154 +/- 28 micrograms/L after 3 and 4 h, respectively. In conclusion, the somatostatin analog octreotide induces IGFBP-1 independently of GH and insulin. As IGFBP-1 regulates the action of IGF-I, octreotide stimulation of IGFBPs may represent an additional pharmacological mechanism for attenuating the GH-IGF-I axis.


Assuntos
Proteínas de Transporte/sangue , Octreotida/farmacologia , Adolescente , Adulto , Idoso , Jejum , Feminino , Hormônio do Crescimento/sangue , Hormônio do Crescimento/deficiência , Humanos , Insulina/sangue , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina , Masculino , Pessoa de Meia-Idade , Hipófise/fisiologia , Valores de Referência , Somatomedinas/análise
13.
J Clin Endocrinol Metab ; 83(4): 1275-83, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9543156

RESUMO

Leukemia inhibitory factor (LIF) exhibits multiple biological activities in various tissues, and we have shown that LIF activates POMC gene transcription in response to immune signals. As higher serum levels of LIF have been reported in septicemia, we measured LIF values in biological fluids by RIA. Immunoreactive LIF was detected in 303 of 428 human serum samples. Circulating LIF detection rates were 69% in acute inflammatory diseases, 83% in chronic inflammatory diseases, 61% in noninflammatory diseases, and 90% in cancer patients. Serum concentrations of human LIF was higher in patients with inflammatory disease than in noninflammatory disease (0.80 +/- 0.10 vs. 0.53 +/- 0.02 ng/mL; P < 0.05) or in cancer patients (0.44 +/- 0.06; P < 0.05). Higher serum human LIF levels were found in septicemia (0.78 +/- 0.14 ng/mL), pneumonia (0.80 +/- 0.10 ng/mL), acute bronchitis (0.88 +/- 0.09 ng/mL), other infections (1.01 +/- 0.17 ng/mL), and systemic lupus erythematosus (SLE; 0.79 +/- 0.06 ng/mL). In 7 septicemia patients, Gram-negative infection was associated with higher LIF levels (1.06 +/- 0.16 ng/mL) than was Gram-positive infection (0.58 +/- 0.14 ng/mL). In patients with acute inflammatory disease, serum LIF levels decreased within several days after hospitalization. To test circulating mouse (m) LIF changes in response to inflammatory stress, lipopolysaccharide (LPS) was injected ip to mice. LPS increased serum mLIF values concordantly with ACTH levels. After i.p. injection of 80 microg LPS, serum mLIF increased by 144% (P < 0.05), 173% (P < 0.05), and 134% at 30, 90, and 120 min respectively. In vitro, however, LPS did not increase ACTH and mLIF secretion from dispersed mouse primary pituitary cells. These results suggest that LIF is an important participant in the pathogenesis of the acute inflammatory response. The elevated serum LIF levels observed in inflammation do not appear to originate from the pituitary.


Assuntos
Inibidores do Crescimento/análise , Interleucina-6 , Linfocinas/análise , Animais , Bioensaio , Células Cultivadas , Meios de Cultura , Ensaio de Imunoadsorção Enzimática , Inibidores do Crescimento/sangue , Humanos , Fator Inibidor de Leucemia , Linfocinas/sangue , Camundongos , Radioimunoensaio , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
14.
J Clin Endocrinol Metab ; 84(8): 2883-7, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10443695

RESUMO

Several cytokines regulate thyroid function and may be involved in the pathogenesis of thyroid disorders, including euthyroid sick syndrome. Leukemia inhibitory factor (LIF), a neuroimmune pleiotropic cytokine, was measured to assess its role in hypothalamic-pituitary-thyroid function. Mean circulating serum LIF levels in 10 hypothyroid patients [TSH, 23+/-0.5 mIU/L (mean+/-SEM); free T4, 0.77+/-0.1 ng/dL] was 0.29+/-0.04 ng/mL, 145% higher (P < 0.04) than in 20 normal subjects (LIF, 0.20+/-0.02 ng/mL; TSH, 2.23+/-0.21 mIU/L; free T4, 1.23+/-0.04 ng/dL) but was not different from those in 10 hyperthyroid patients (LIF, 0.21+/-0.03 ng/mL; TSH, 0.01+/-0.00 mIU/L; free T4, 3.63+/-0.51 ng/dL). Serum LIF concentrations linearly correlated with serum TSH in the 40 samples (r = 0.58, P < 0.001). When T4 (1-8 microg/kg x day) was administered to cynomolgus monkeys with methimazole-induced hypothyroidism, serum T4 and T3 levels increased appropriately, and TSH and LIF concentrations decreased. When methimazole was given alone, both serum TSH (146+/-30 mIU/L) and LIF (8.84+/-0.49 ng/mL) were markedly induced. When methimazole together with T4 (>2 microg/kg x day) was administered, both serum TSH (7.5+/-1.2 mIU/L) and LIF (6.22+/-0.31 ng/mL) were lowered (P < 0.01). Monkey serum LIF levels and log TSH levels also correlated (r = 0.72, P < 0.01). Cultured thyroid carcinoma cells produced LIF (9.2 ng/10(6) cells/48 h). TSH (100 mIU/mL) and interleukin (IL)-6 (10 nmol/L) stimulated in vitro LIF secretion from the cells by 170+/-12% (P < 0.05) and 261+/-8% (P < 0.05), respectively. Dexamethasone (1 micromol/L) inhibited basal LIF concentration by 83% (P < 0.05), whereas TSH and IL-6 stimulated LIF by 52% (P = 0.04) and 42% (P = 0.03), respectively. However, using Northern blot analysis, we could not observe induction of LIF mRNA by TSH, suggesting that LIF regulation by TSH may be due to stimulation of secretion. The results show that the thyroid gland is a source of LIF production; TSH, IL-6, and glucocorticoid influence thyroid cell LIF expression. The correlation between TSH and LIF suggests that LIF may participate in the physiologic regulation of hypothalamic-pituitary-thyroid function.


Assuntos
Inibidores do Crescimento/biossíntese , Hipotireoidismo/diagnóstico , Linfocinas/biossíntese , Glândula Tireoide/metabolismo , Animais , Biomarcadores , Feminino , Inibidores do Crescimento/sangue , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Interleucina-6/farmacologia , Fator Inibidor de Leucemia , Linfocinas/sangue , Macaca fascicularis , Masculino , Tireotropina/sangue
15.
J Clin Endocrinol Metab ; 66(5): 1010-3, 1988 May.
Artigo em Inglês | MEDLINE | ID: mdl-3360894

RESUMO

To investigate the dose-response relationship between thyroid hormone and linear growth, we studied 10 castrated prepubertal cynomolgus monkeys. Hypothyroidism was induced by administration of methimazole (0.0125% in drinking water) and was confirmed by high serum TSH levels (greater than 40 mU/L) in all animals. Subsequently, each animal received 1, 2, 4, or 8 micrograms/kg.day T4, im, for 9 weeks. The sequence of T4 doses was random, and 6 weeks elapsed between successive T4 doses. Serum T4, T3, TSH, and insulin-like growth factor I (IGF-I) levels and lower leg length were measured every 3 weeks. Methimazole administration decreased thyroid hormone and IGF-I levels and lower leg growth rate. With increasing doses of exogenous T4, serum T4, T3, and IGF-I as well as lower leg growth rate increased significantly. Animals not given T4 had a 65% decrease in lower leg growth rate (P less than 0.01). Animals given 4 and 8 micrograms/kg.day T4 had 56% and 73% increases, respectively, in lower leg growth rate compared to baseline (P less than 0.05 and P less than 0.01, respectively). Lower leg growth rate correlated better with serum T3 (r = 0.50; P less than 0.001) than with serum T4 (r = 0.29; P less than 0.05). Lower leg growth rate also correlated with serum IGF-I levels (r = 0.53; P less than 0.001). Serum IGF-I correlated with serum T3 (r = 0.47; P less than 0.001), but not with serum T4. We conclude that increased serum T4 and T3 levels cause progressive increases in growth velocity and IGF-I levels over a range from moderate hypothyroidism to moderate hyperthyroidism. Growth velocity and IGF-I levels correlated more strongly with the serum T3 than with the serum T4 level.


Assuntos
Crescimento/efeitos dos fármacos , Hormônios Tireóideos/administração & dosagem , Animais , Castração , Relação Dose-Resposta a Droga , Feminino , Hipertireoidismo/sangue , Hipotireoidismo/sangue , Hipotireoidismo/induzido quimicamente , Macaca fascicularis , Masculino , Metimazol/administração & dosagem , Somatomedinas/sangue , Tiroxina/administração & dosagem , Tri-Iodotironina/administração & dosagem
16.
J Clin Endocrinol Metab ; 73(2): 314-7, 1991 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-1856262

RESUMO

GH-releasing peptide (GHRP; His-D-Trp-Ala-Trp-D-Phe-Lys-NH2), a hexapeptide derived from enkephalin, has been shown to have GH-releasing activity in man and several animal species. To characterize the GHRP dose-response curve and compare it with that of GH-releasing hormone [GHRH-(1-44)NH2], six unanesthetized young adult cynomolgus macaques were tested with a range of iv doses of GHRP or GHRH in random order. Animals were fitted with vests and tethers. Blood samples were obtained before and at 15-min intervals after the administration of drugs. Doses ranged from 0.03-3 mg/kg for GHRP and from 1-30 micrograms/kg for GHRH. The dose-response curves for the two peptides were not parallel. GHRP had lower potency, but evoked a much higher peak GH response than GHRH (greater than 55 vs. 12 micrograms/L). Because one of the proposed mechanisms of action of GHRP is the inhibition of somatostatin (SS), we tested the effects of propranolol, which inhibits SS, on the GH responses to GHRH and GHRP. Propranolol was given at a dose of 14 micrograms/kg, iv, 10 min before the injection of saline, GHRH (10 micrograms/kg), or GHRP (1 mg/kg). GH responses to propranolol alone did not differ from those to placebo (peak, 6 +/- 2 vs. 8 +/- 2 micrograms/L). However, propranolol pretreatment doubled the GH responses to both GHRH and GHRP compared with those to GHRH or GHRP alone 28 +/- 5 micrograms/L vs. 14 +/- 5 (P less than 0.05) and 54 +/- 2 vs. 25 +/- 6 micrograms/L (P less than 0.001), respectively]. These results show that GHRP causes a potent dose-dependent release of GH in this primate species. Since GHRP can produce a greater maximal GH response than GHRH, mechanisms other than release of endogenous GHRH must be involved.


Assuntos
Hormônio Liberador da Corticotropina/farmacologia , Hormônio do Crescimento/metabolismo , Oligopeptídeos/farmacologia , Animais , Relação Dose-Resposta a Droga , Hormônio do Crescimento/sangue , Macaca fascicularis , Masculino , Propranolol/farmacologia , Valores de Referência
17.
J Endocrinol ; 174(3): 379-86, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12208657

RESUMO

Proteasome inhibitors induce apoptosis in some malignant cells, and we show here that these inhibitors induce apoptosis in rat pituitary MMQ and GH3 tumor cells but not in normal pituitary cells. Three proteasome inhibitors, PSI, MG-132, and lactacystin, but not the calpain inhibitor, ALLM, dose- and time-dependently caused apoptosis in these cells, and 10 microM PSI caused apoptosis in 70% of MMQ cells and in 25% of GH3 cells within 24 h. A lower PSI dose (10 nM) inhibited GH3 cell growth without causing significant apoptosis or affecting prolactin secretion. Primary rat pituitary cells were resistant to both PSI and MG-132 and did not undergo apoptosis. In MMQ cells, DNA synthesis was slowed (approximately 30%) after 6 h of 10 microM PSI treatment and a partial cell cycle block at G2/M was evident after 8 h. Colorimetric caspase substrate assay and Western blotting of caspase substrates showed that caspases 2 and 3 are activated by PSI while caspases 6 and 8 remained inactive. A broad-range caspase inhibitor, caspase inhibitor III, prevented apoptosis induced by PSI. The results show that proteasome inhibitors induce apoptosis in rat pituitary tumor cells by specific caspase activation. This novel group of drugs may potentially be used in treatment of aggressive pituitary tumors, especially as their action appears relative for tumor cells.


Assuntos
Acetilcisteína/análogos & derivados , Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Complexos Multienzimáticos/antagonistas & inibidores , Neoplasias Hipofisárias/patologia , Acetilcisteína/farmacologia , Animais , Western Blotting/métodos , Caspases/metabolismo , Células Cultivadas , Cisteína Endopeptidases , Ativação Enzimática/efeitos dos fármacos , Feminino , Hormônio do Crescimento/metabolismo , Marcação In Situ das Extremidades Cortadas , Leupeptinas/farmacologia , Adeno-Hipófise/citologia , Adeno-Hipófise/efeitos dos fármacos , Neoplasias Hipofisárias/metabolismo , Prolactinoma/patologia , Complexo de Endopeptidases do Proteassoma , Ratos , Células Tumorais Cultivadas
18.
Ann N Y Acad Sci ; 840: 162-73, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9629248

RESUMO

Leukemia inhibitory factor (LIF), a pleiotropic cytokine, is expressed in both fetal and adult pituitary tissue, and LIF immunoreactivity is found in functional human pituitary tumors. LIF induces basal, and augments CRH-induced, POMC mRNA and ACTH secretion from AtT20 cells. Therefore, we examined LIF signaling and LIF regulation of POMC expression in AtT20 cells. Immunoneutralization studies demonstrated the dependence of LIF action on both the specific LIF receptor (35% inhibition; p < 0.05) and also the gpl30 affinity converter (41% inhibition; p < 0.05). These antisera also attenuate basal ACTH secretion without added LIF. LIF rapidly induced tyrosyl phosphorylation of both STAT 1 alpha, and STAT beta and also induced phosphorylation of a novel STAT 1 alpha related protein p115. LIF induced POMC transcription (-706/+64) and strikingly potentiated CRH action (up to 18-fold induction). This synergy involved cAMP-dependent pathways, as forskolin action was also potentiated by LIF. Deletion of the major CRH-responsive region in POMC (-323/-166) abolished both CRH and LIF action on POMC transcription. Thus LIF action in pituitary corticotrophs is dependent on LIF receptor heterodimerisation with gpl30 and involves STAT protein tyrosyl phosphorylation. LIF enhances POMC transcription and strongly potentiates the well-documented action of CRH on the POMC gene. These results define a subcellular mechanism for an immuno-neuroendocrine interface between peripheral afferent signals and the HPA axis.


Assuntos
Inibidores do Crescimento/fisiologia , Interleucina-6 , Linfocinas/fisiologia , Pró-Opiomelanocortina/genética , Transcrição Gênica/fisiologia , Hormônio Adrenocorticotrópico/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Inibidores do Crescimento/farmacologia , Fator Inibidor de Leucemia , Subunidade alfa de Receptor de Fator Inibidor de Leucemia , Linfocinas/farmacologia , Proteínas de Membrana Lisossomal , Glicoproteínas de Membrana/fisiologia , Camundongos , Fosforilação , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Pró-Opiomelanocortina/efeitos dos fármacos , Receptores de Citocinas/fisiologia , Receptores de OSM-LIF , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Transativadores/metabolismo , Transcrição Gênica/efeitos dos fármacos
19.
Fertil Steril ; 54(3): 532-4, 1990 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1697814

RESUMO

Measurements of the serum concentrations of the endometrial proteins IGFBP-1 and PP14 were made in an attempt to monitor the adequacy of the P effect in women receiving 1 year of 100 to 200 micrograms of percutaneous E2 for 28 days each month and 10 mg oral MPA given during the last 12 days of E2 administration. There were no significant changes in IGFBP-1 levels; a small, but significant increase in PP14 levels after E2 plus MPA was noted, but there was substantial overlap between basal and postprogestogen therapy serum PP14 concentrations. Serum concentrations of PP14, but not IGFBP-1, were slightly higher in women whose endometrial biopsies demonstrated a P effect, but again, there was substantial overlap with the values found in women whose biopsies showed only an estrogen effect. Serum measurements of IGFBP-1 and PP14 are not useful for monitoring postmenopausal replacement therapy with percutaneous E2 and oral MPA.


Assuntos
Proteínas de Transporte/sangue , Estradiol/uso terapêutico , Terapia de Reposição de Estrogênios/métodos , Glicoproteínas , Menopausa/sangue , Proteínas da Gravidez/sangue , Progesterona/uso terapêutico , Adulto , Idoso , Proteínas de Transporte/imunologia , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Feminino , Glicodelina , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Pessoa de Meia-Idade , Proteínas da Gravidez/imunologia , Progesterona/fisiologia , Radioimunoensaio
20.
Heart ; 77(3): 276-9, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9093050

RESUMO

OBJECTIVES: To study the feasibility of congenital ventricular septal defect occlusion by the buttoned device and to establish guidelines for its safe and effective application. DESIGN: A descriptive study of all patients with a congenital ventricular septal defect undergoing transcatheter occlusion with the buttoned device, from March 1994 to May 1995. These patients were otherwise candidates for elective surgery at their institutions because they had persistence of a significant shunt (Qp:Qs = 1.5-2.1:1, median = 1.7), with left ventricular enlargement and/or symptoms, although their systolic pulmonary artery pressure was invariably normal (20-28 mm Hg, median = 25). The angiographic diameter of the defect ranged from 2.5 to 14 mm (median 6 mm). SETTING: A multi-institutional study. PATIENTS: Out of 25 cases attempted, 18 children and adults aged 4-35 years had devices implanted. Fifteen of these patients had membranous ventricular septal defects and three had muscular defects. All patients with a membranous ventricular septal defect had an associated aneurysm of the membranous septum. INTERVENTIONS: The buttoned device was introduced either directly or, in the last 12 cases, over a wire bridging the femoral artery and the femoral or jugular vein; the devices were delivered through 7-9 French (F) long sheaths. A membranous defect was regarded as suitable for device closure if the distance from the centre of the defect to the insertion of the right coronary aortic valve leaflet was more than 50% of the size of the required device. The device was guided by echocardiography and fluoroscopy. All muscular defects were corrected through the right jugular vein and all membranous ones through the femoral vein. RESULTS: All 18 patients underwent initial successful implantation of the device. In thirteen patients the shunts were completely occluded and in the remaining five there were trivial residual shunts. In two patients with membranous ventricular septal defects a change from the original position was noticed at two weeks; mild aortic regurgitation developed in one and the murmur recurred in the other; the devices had to be removed surgically. One patient developed transient third degree atrioventricular block during implantation; no tricuspid regurgitation was observed. CONCLUSION: Clinical occlusion of congenital ventricular septal defects was achieved in 16 out of the 18 attempted cases (13 full occlusions). Membranous ventricular septal defect occlusion can be effective and safe if patients and device sizes are carefully selected.


Assuntos
Cateterismo Cardíaco , Comunicação Interventricular/terapia , Adolescente , Adulto , Criança , Pré-Escolar , Ecocardiografia , Seguimentos , Humanos , Pessoa de Meia-Idade , Equipamentos Cirúrgicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA