Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 246
Filtrar
1.
Trends Endocrinol Metab ; 32(6): 338-340, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33771415

RESUMO

Growth hormone releasing hormone (GHRH) is the integral regulator of the growth hormone (GH)-insulin-like growth factor 1 (IGF-1) axis. It exerts mitogenic effects in a plethora of progressive cancers. Recent evidence suggests the emerging role of that 44-amino acid (aa) neuropeptide in lung endothelial barrier function (EBF), which will be discussed herein.


Assuntos
Endotélio/fisiologia , Hormônio Liberador de Hormônio do Crescimento , Hormônio do Crescimento Humano , Fator de Crescimento Insulin-Like I , Pulmão/fisiologia , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Humanos
2.
J Neurosci ; 40(49): 9455-9466, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33158965

RESUMO

Gonadal steroids modulate growth hormone (GH) secretion and the pubertal growth spurt via undefined central pathways. GH-releasing hormone (GHRH) neurons express estrogen receptor α (ERα) and androgen receptor (AR), suggesting changing levels of gonadal steroids during puberty directly modulate the somatotropic axis. We generated mice with deletion of ERα in GHRH cells (GHRHΔERα), which displayed reduced body length in both sexes. Timing of puberty onset was similar in both groups, but puberty completion was delayed in GHRHΔERα females. Lack of AR in GHRH cells (GHRHΔAR mice) induced no changes in body length, but puberty completion was also delayed in females. Using a mouse model with two reporter genes, we observed that, while GHRHtdTom neurons minimally colocalize with Kiss1hrGFP in prepubertal mice, ∼30% of GHRH neurons coexpressed both reporter genes in adult females, but not in males. Developmental analysis of Ghrh and Kiss1 expression suggested that a subpopulation of ERα neurons in the arcuate nucleus of female mice undergoes a shift in phenotype, from GHRH to Kiss1, during pubertal transition. Our findings demonstrate that direct actions of gonadal steroids in GHRH neurons modulate growth and puberty and indicate that GHRH/Kiss1 dual-phenotype neurons play a sex-specific role in the crosstalk between the somatotropic and gonadotropic axes during pubertal transition.SIGNIFICANCE STATEMENT Late maturing adolescents usually show delayed growth and bone age. At puberty, gonadal steroids have stimulatory effects on the activation of growth and reproductive axes, but the existence of gonadal steroid-sensitive neuronal crosstalk remains undefined. Moreover, the neural basis for the sex differences observed in the clinical arena is unknown. Lack of ERα in GHRH neurons disrupts growth in both sexes and causes pubertal delay in females. Deletion of androgen receptor in GHRH neurons only delayed female puberty. In adult females, not males, a subset of GHRH neurons shift phenotype to start producing Kiss1. Thus, direct estrogen action in GHRH/Kiss1 dual-phenotype neurons modulates growth and puberty and may orchestrate the sex differences in endocrine function observed during pubertal transition.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Crescimento/fisiologia , Kisspeptinas/fisiologia , Maturidade Sexual/fisiologia , Transdução de Sinais/fisiologia , Animais , Receptor alfa de Estrogênio/genética , Feminino , Hormônios Esteroides Gonadais/sangue , Hormônios Esteroides Gonadais/fisiologia , Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/genética , Hipotálamo/metabolismo , Kisspeptinas/genética , Masculino , Camundongos , Camundongos Knockout , Receptores Androgênicos/fisiologia , Caracteres Sexuais , Maturidade Sexual/genética , Transdução de Sinais/genética
3.
Peptides ; 86: 153-161, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27816751

RESUMO

Triple-negative breast cancer (TNBC) is a subset of breast cancers which is negative for expression of estrogen and progesterone receptors and human epidermal growth factor receptor-2 (HER2). Chemotherapy is currently the only form of treatment for women with TNBC. Growth hormone-releasing hormone (GHRH) and epidermal growth factor (EGF) are autocrine/paracrine growth factors in breast cancer and a substantial proportion of TNBC expresses receptors for GHRH and EGF. The aim of this study was to evaluate the interrelationship between both these signaling pathways in MDA-MB-468 human TNBC cells. We evaluated by Western blot assays the effect of GHRH on transactivation of EGF receptor (EGFR) as well as the elements implicated. We assessed the effect of GHRH on migration capability of MDA-MB-468 cells as well as the involvement of EGFR in this process by means of wound-healing assays. Our findings demonstrate that in MDA-MB-468 cells the stimulatory activity of GHRH on tyrosine phosphorylation of EGFR is exerted by two different molecular mechanisms: i) through GHRH receptors, GHRH stimulates a ligand-independent activation of EGFR involving at least cAMP/PKA and Src family signaling pathways; ii) GHRH also stimulates a ligand-dependent activation of EGFR implicating an extracellular pathway with an important role for metalloproteinases. The cross-talk between EGFR and GHRHR may be impeded by combining drugs acting upon GHRH receptors and EGFR family members. This combination of GHRH receptors antagonists with inhibitors of EGFR signalling could enhance the efficacy of both types of agents as well as reduce their doses increasing therapeutic benefits in management of human breast cancer.


Assuntos
Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Ativação Transcricional , Neoplasias de Mama Triplo Negativas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Receptores ErbB/metabolismo , Feminino , Humanos , Metaloproteinases da Matriz/metabolismo , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia , Quinases da Família src/metabolismo
4.
Endocrinology ; 156(9): 3239-52, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26110916

RESUMO

Skeletal muscle atrophy is a consequence of different chronic diseases, including cancer, heart failure, and diabetes, and also occurs in aging and genetic myopathies. It results from an imbalance between anabolic and catabolic processes, and inflammatory cytokines, such as TNF-α, have been found elevated in muscle atrophy and implicated in its pathogenesis. GHRH, in addition to stimulating GH secretion from the pituitary, exerts survival and antiapoptotic effects in different cell types. Moreover, we and others have recently shown that GHRH displays antiapoptotic effects in isolated cardiac myocytes and protects the isolated heart from ischemia/reperfusion injury and myocardial infarction in vivo. On these bases, we investigated the effects of GHRH on survival and apoptosis of TNF-α-treated C2C12 myotubes along with the underlying mechanisms. GHRH increased myotube survival and prevented TNF-α-induced apoptosis through GHRH receptor-mediated mechanisms. These effects involved activation of phosphoinositide 3-kinase/Akt pathway and inactivation of glycogen synthase kinase-3ß, whereas mammalian target of rapamycin was unaffected. GHRH also increased the expression of myosin heavy chain and the myogenic transcription factor myogenin, which were both reduced by the cytokine. Furthermore, GHRH inhibited TNF-α-induced expression of nuclear factor-κB, calpain, and muscle ring finger1, which are all involved in muscle protein degradation. In summary, these results indicate that GHRH exerts survival and antiapoptotic effects in skeletal muscle cells through the activation of anabolic pathways and the inhibition of proteolytic routes. Overall, our findings suggest a novel therapeutic role for GHRH in the treatment of muscle atrophy-associated diseases.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Animais , Apoptose , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Camundongos , Fibras Musculares Esqueléticas/citologia , Proteínas Musculares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteólise , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Fator de Necrose Tumoral alfa
5.
Elife ; 2: e01098, 2013 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-24175087

RESUMO

We examine the impact of targeted disruption of growth hormone-releasing hormone (GHRH) in mice on longevity and the putative mechanisms of delayed aging. GHRH knockout mice are remarkably long-lived, exhibiting major shifts in the expression of genes related to xenobiotic detoxification, stress resistance, and insulin signaling. These mutant mice also have increased adiponectin levels and alterations in glucose homeostasis consistent with the removal of the counter-insulin effects of growth hormone. While these effects overlap with those of caloric restriction, we show that the effects of caloric restriction (CR) and the GHRH mutation are additive, with lifespan of GHRH-KO mutants further increased by CR. We conclude that GHRH-KO mice feature perturbations in a network of signaling pathways related to stress resistance, metabolic control and inflammation, and therefore provide a new model that can be used to explore links between GHRH repression, downregulation of the somatotropic axis, and extended longevity. DOI:http://dx.doi.org/10.7554/eLife.01098.001.


Assuntos
Restrição Calórica , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Longevidade , Animais , Perfilação da Expressão Gênica , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/uso terapêutico , Hipopituitarismo/tratamento farmacológico , Fígado/metabolismo , Camundongos , Camundongos Knockout , Fator 2 Relacionado a NF-E2/metabolismo , Fenótipo , Xenobióticos/metabolismo
6.
Praxis (Bern 1994) ; 102(13): 777-84, 2013 Jun 19.
Artigo em Alemão | MEDLINE | ID: mdl-23773936

RESUMO

Growth Hormone therapy has been used therapeutically for over 50 years. Until recently, growth hormone therapy has been restricted for children and adolescents with proven hypothalamic-pituitary short stature. Today some other causes - but not all - can be treated with growth hormone. To the well-established indications belong apart from proven growth hormone deficiency, children with Turner Syndrome and with Prader Willi Syndrome, children born small for gestational age without catch-up growth and children with chronic kidney disease and with some haematological and oncological diseases. Careful and accurate diagnosis is essential. Growth hormone therapy is rare in everyday practice and requires close cooperation with a pediatric endocrinologist.


L'hormone de croissance est utilisée depuis 50 ans à des fins thérapeutiques. Jusque à récemment seule une petite taille causée par une atteinte de l'axe hypothalamo-hypophysaire était une indication à un traitement par hormone de croissance. Actuellement, des étiologies variées de petite taille peuvent être traitées par hormone de croissance. Parmi les indications, on peut citer un déficit en hormone de croissance, les syndromes de Turner et de Prader-Willi, un retard de croissance intra-utérin sans rattrapage de la taille, et aussi une insuffisance rénale ou certaines maladies hématologiques ou oncologiques. Avant le traitement un diagnostic précis est essentiel. Les différentes indications qui peuvent amener à un traitement sont rares dans la pratique quotidienne, il est donc important de favoriser une prise en charge commune avec l'endocrinologue pédiatre.


Assuntos
Transtornos do Crescimento/tratamento farmacológico , Hormônio do Crescimento Humano/uso terapêutico , Adolescente , Estatura/efeitos dos fármacos , Estatura/fisiologia , Criança , Comportamento Cooperativo , Transtornos do Crescimento/etiologia , Transtornos do Crescimento/fisiopatologia , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Hormônio do Crescimento Humano/efeitos adversos , Hormônio do Crescimento Humano/fisiologia , Humanos , Fator de Crescimento Insulin-Like I/fisiologia , Comunicação Interdisciplinar , Valores de Referência , Fatores de Risco
7.
Peptides ; 37(1): 63-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22819774

RESUMO

Lung cancers which show increased vascularization and high microvessel density are considered highly metastatic and with poor prognosis. Growth hormone releasing hormone (GHRH) antagonists are anticancer agents without adverse events in lung cancer tumor models. In the present study we investigated the in vitro effect of GHRH antagonist, MZ-5-156, on focal adhesion kinase (FAK) activity, on the expression of MMP-2 and MMP-9 metalloproteinases, as well as on vascular endothelial growth factor (VEGF) levels in A549 non-small cell lung (NSCLC) cancer cells and H727 bronchial carcinoid cells. We demonstrate for the first time that GHRH antagonist, MZ-5-156, inhibits FAK signaling in lung cancer cells and decreases the expression of additional factors involved in angiogenesis and invasion. In contrast, GHRH itself counteracted these effects. Our study contributes to the further understanding of the processes which govern the mechanism of action of GHRH and its antagonists in cancers.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Hormônio Liberador de Hormônio do Crescimento/antagonistas & inibidores , Sermorelina/análogos & derivados , Fator A de Crescimento do Endotélio Vascular/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Expressão Gênica , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Humanos , Neoplasias Pulmonares , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Sermorelina/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética
8.
Am J Physiol Regul Integr Comp Physiol ; 301(4): R1143-52, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21795635

RESUMO

Although stimulatory (feedforward) and inhibitory (feedback) dynamics jointly control neurohormone secretion, the factors that supervise feedback restraint are poorly understood. To parse the regulation of growth hormone (GH) escape from negative feedback, 25 healthy men and women were studied eight times each during an experimental GH feedback clamp. The clamp comprised combined bolus infusion of GH or saline and continuous stimulation by saline GH-releasing hormone (GHRH), GHRP-2, or both peptides after randomly ordered supplementation with placebo (both sexes) vs. E(2) (estrogen; women) and T (testosterone; men). Endpoints were GH pulsatility and entropy (a model-free measure of feedback quenching). Gender determined recovery of pulsatile GH secretion from negative feedback in all four secretagog regimens (0.003 ≤ P ≤ 0.017 for women>men). Peptidyl secretagog controlled the mass, number, and duration of feedback-inhibited GH secretory bursts (each, P < 0.001). E(2)/T administration potentiated both pulsatile (P = 0.006) and entropic (P < 0.001) modes of GH recovery. IGF-I positively predicted the escape of GH secretory burst number and mode (P = 0.022), whereas body mass index negatively forecast GH secretory burst number and mass (P = 0.005). The composite of gender, body mass index, E(2), IGF-I, and peptidyl secretagog strongly regulates the escape of pulsatile and entropic GH secretion from autonegative feedback. The ensemble factors identified in this preclinical investigation enlarge the dynamic model of GH control in humans.


Assuntos
Índice de Massa Corporal , Estradiol/fisiologia , Retroalimentação Fisiológica/fisiologia , Hormônio do Crescimento/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Idoso , Estudos Cross-Over , Método Duplo-Cego , Estradiol/farmacologia , Retroalimentação Fisiológica/efeitos dos fármacos , Feminino , Hormônio do Crescimento/farmacologia , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Masculino , Pessoa de Meia-Idade , Caracteres Sexuais , Testosterona/farmacologia , Testosterona/fisiologia
9.
Eur Respir J ; 38(4): 870-7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21406516

RESUMO

Upper airway obstruction (UAO) can lead to abnormal growth hormone (GH) homeostasis and growth retardation but the mechanisms are unclear. We explored the effect of UAO on hypothalamic GH-releasing hormone (GHRH), which has a role in both sleep and GH regulation. The tracheae of 22-day-old rats were narrowed; UAO and sham-operated animals were sacrificed 16 days post-surgery. To stimulate slow-wave sleep (SWS) and GH secretion, rats were treated with ritanserin (5-HT(2) receptor antagonist). Sleep was measured with a telemetric system. Hypothalamic GHRH, hypothalamic GHRH receptor (GHRHR) and GH receptor, and orexin were analysed using ELISA, real-time PCR and Western blot. UAO decreased hypothalamic GHRH, GHRHR and GH receptor levels, while orexin mRNA increased (p<0.01). In UAO rats, the duration of wakefulness was elevated and the duration of SWS, paradoxical sleep and slow-wave activity was reduced (p<0.001). Ritanserin alleviated these effects, i.e. normalised hypothalamic GHRH content, decreased wake duration, increased duration and depth of SWS, and attenuated growth impairment (p<0.001). Here, we present evidence that growth retardation in UAO is associated with a reduction in hypothalamic GHRH content. Our findings show that abnormalities in the GHRH/GH axis underlie both growth retardation and SWS-disorder UAO.


Assuntos
Transtornos do Crescimento/fisiopatologia , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Hipotálamo/fisiologia , Apneia Obstrutiva do Sono/fisiopatologia , Sono/fisiologia , Animais , Modelos Animais de Doenças , Transtornos do Crescimento/metabolismo , Hormônio do Crescimento/sangue , Hormônio Liberador de Hormônio do Crescimento/genética , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/sangue , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Neuropeptídeos/sangue , Neuropeptídeos/genética , Orexinas , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Ritanserina/farmacologia , Antagonistas da Serotonina/farmacologia , Sono/efeitos dos fármacos , Apneia Obstrutiva do Sono/metabolismo , Telemetria
10.
Proc Natl Acad Sci U S A ; 107(43): 18611-5, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-20937882

RESUMO

Despite the well-documented action of growth hormone-releasing hormone (GHRH) on the stimulation of production and release of growth hormone (GH), the effects of GHRH in peripheral tissues are incompletely explored. In this study, we show that GHRH plays a role in wound healing and tissue repair by acting primarily on wound-associated fibroblasts. Mouse embryonic fibroblasts (MEFs) in culture and wound-associated fibroblasts in mice expressed a splice variant of the receptors for GHRH (SV1). Exposure of MEFs to 100 nM and 500 nM GHRH or the GHRH agonist JI-38 stimulated the expression of α-smooth muscle actin (αSMA) based on immunoblot analyses as well as the expression of an αSMA-ß-galactosidase reporter transgene in primary cultures of fibroblasts isolated from transgenic mice. Consistent with this induction of αSMA expression, results of transwell-based migration assays and in vitro wound healing (scratch) assays showed that both GHRH and GHRH agonist JI-38 stimulated the migration of MEFs in vitro. In vivo, local application of GHRH or JI-38 accelerated healing in skin wounds of mice. Histological evaluation of skin biopsies showed that wounds treated with GHRH and JI-38 were both characterized by increased abundance of fibroblasts during the early stages of wound healing and accelerated reformation of the covering epithelium at later stages. These results identify another function of GHRH in promoting skin tissue wound healing and repair. Our findings suggest that GHRH may have clinical utility for augmenting healing of skin wounds resulting from trauma, surgery, or disease.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/agonistas , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Cicatrização/efeitos dos fármacos , Actinas/genética , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Hormônio do Crescimento/análogos & derivados , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cicatrização/genética , Cicatrização/fisiologia
12.
Front Horm Res ; 38: 206-211, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20616513

RESUMO

Ghrelin, a 28-amino-acid octanoylated peptide predominantly produced by the stomach, was discovered to be the natural ligand of the type 1a GH secretagogue receptor. Thus, it was considered as a natural GH secretagogue (GHS) additional to GHRH, although later on ghrelin has mostly been considered a major orexigenic factor. The GH-releasing action of ghrelin takes place both directly on pituitary cells and through modulation of GHRH from the hypothalamus; some functional anti-somatostatin action has also been shown. However, even at the neuroendocrine level, ghrelin is much more than a natural GHS. In fact, it significantly stimulates prolactin secretion in humans, independent of both gender and age and probably involving a direct action on somatomammotroph cells. Above all, ghrelin and synthetic GHS possess an acute stimulatory effect on the activity of the hypothalamus-pituitary-adrenal axis in humans, which is, at least, similar to that of the opioid antagonist naloxone, arginine vasopressin and even corticotropin-releasing hormone. Also, ghrelin plays a relevant role in the modulation of the hypothalamic-pituitary-gonadal function, with a predominantly CNS-mediated inhibitory effect upon the gonadotropin pulsatility both in animals and in humans.


Assuntos
Grelina/fisiologia , Adeno-Hipófise/fisiologia , Hormônio Adrenocorticotrópico/metabolismo , Hormônio Liberador da Corticotropina/fisiologia , Gonadotropinas/metabolismo , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Humanos , Prolactina/fisiologia
13.
Am J Physiol Regul Integr Comp Physiol ; 298(1): R147-56, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19889861

RESUMO

We examined whether growth hormone-releasing hormone (GHRH) may promote non-rapid eye movement (NREM) sleep via activation of GABAergic neurons in the preoptic area. Male Sprague-Dawley rats were implanted with EEG, EMG electrodes and a unilateral intracerebroventricular cannula. Groups of rats received injections (3 microl icv) with gonadotropin-releasing hormone (GHRH) (0.1 nmol/100 g body wt) or equal volume of physiological saline at the onset of the dark period and were permitted spontaneous sleep for 90 min. Separate groups of rats were sleep deprived by gentle handling for 90 min, beginning at the time of GHRH or saline injection, at the onset of the dark period. Other groups of rats received intracerebroventricular octreotide (somatostatin analog OCT) injections, intracerebroventricular injection of one of two doses of competitive GHRH antagonist, or intracerebroventricular saline injection at light onset and were then permitted 90 min spontaneous sleep-waking. Rats were killed immediately after the 90-min sleep/wake monitoring period. Brain tissue was processed for immunohistochemistry for c-Fos protein and glutamic acid decarboxylase (GAD). Single c-Fos and dual Fos-GAD cell counts were determined in the median preoptic nucleus (MnPN), and in the core and the extended parts of the ventrolateral preoptic nucleus (cVLPO and exVLPO). Intracerebroventricular GHRH elicited a significant increase in NREM sleep amount. Double-labeled Fos+GAD cell counts were significantly elevated after GHRH injection in the MnPN and VLPO in both undisturbed and sleep-deprived groups. OCT and GHRH antagonist significantly decreased NREM sleep amount compared with control rats. OCT injection increased single c-Fos-labeled cell counts in the MnPN, but not in the VLPO. Double-labeled cell counts were significantly reduced after OCT and the high dose of GHRH antagonist injection in all areas examined. These findings identify GABAergic neurons in the MnPN and VLPO as potential targets of the sleep-regulatory actions of GHRH.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/fisiologia , Neurônios/fisiologia , Área Pré-Óptica/fisiologia , Sono/fisiologia , Animais , Eletroencefalografia , Eletromiografia , Glutamato Descarboxilase/metabolismo , Hormônio Liberador de Hormônio do Crescimento/administração & dosagem , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Injeções Intraventriculares , Masculino , Modelos Animais , Neurônios/efeitos dos fármacos , Octreotida/administração & dosagem , Octreotida/farmacologia , Área Pré-Óptica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley , Sono REM/fisiologia , Somatostatina/análogos & derivados , Ácido gama-Aminobutírico/fisiologia
14.
Eur J Endocrinol ; 161(5): 681-6, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19696160

RESUMO

OBJECTIVE: In Cushing's disease (CD), GH responsiveness to several stimuli, including ghrelin, GHRP-6, and GHRH, is blunted. Recovery of GH secretion after remission of hypercortisolism after transsphenoidal surgery, radiotherapy, or adrenalectomy is controversial. There are no studies evaluating the effect of primary clinical treatment with ketoconazole on GH secretion in CD. The aim of this study is to compare ghrelin-, GHRP-6-, and GHRH-induced GH release before and after ketoconazole in CD. DESIGN: GH responses to ghrelin, GHRP-6, and GHRH of eight untreated patients with CD (mean age: 33.8+/-3.1 years; body mass index: 28.5+/-0.8 kg/m(2)) were evaluated before and after 3 and 6 months of ketoconazole treatment, and compared with 11 controls (32.1+/-2.5; 25.0+/-0.8). Methods Serum GH was measured by an immunofluorometric assay and urinary free cortisol (UFC) by liquid chromatography and tandem mass spectrometry. RESULTS: After ketoconazole use, mean UFC decreased significantly (before: 222.4+/-35.0 microg/24 h; third month: 61.6+/-10.1; sixth month: 39.1+/-10.9). Ghrelin-induced GH secretion increased significantly after 6 months (peak before: 6.8+/-2.3 microg/l; sixth month: 16.0+/-3.6), but remained lower than that of controls (54.1+/-11.2). GH release after GHRP-6 increased, although not significantly, while GH responsiveness to GHRH was unchanged. CONCLUSIONS: Ghrelin-induced GH release increases significantly after 6 months of ketoconazole treatment in CD. This could suggest that a decrease in cortisol levels during this time period can partially restore glucocorticoid-induced GH suppression in CD. GH-releasing mechanisms stimulated by ghrelin/GHS could be more sensitive, as no changes in GHRH-induced GH release were observed.


Assuntos
Grelina/administração & dosagem , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Hormônio do Crescimento Humano/metabolismo , Cetoconazol/administração & dosagem , Oligopeptídeos/administração & dosagem , Hipersecreção Hipofisária de ACTH/tratamento farmacológico , Hipersecreção Hipofisária de ACTH/fisiopatologia , Adulto , Área Sob a Curva , Feminino , Grelina/fisiologia , Hormônio Liberador de Hormônio do Crescimento/administração & dosagem , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hormônio do Crescimento Humano/sangue , Humanos , Hidrocortisona/sangue , Hidrocortisona/urina , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Oligopeptídeos/fisiologia , Hipersecreção Hipofisária de ACTH/metabolismo , Estatísticas não Paramétricas , Adulto Jovem
15.
Domest Anim Endocrinol ; 37(3): 181-7, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19616401

RESUMO

Estradiol increases basal growth hormone (GH) concentrations in sheep and cattle. This study sought to determine the effects of estradiol on GH-releasing hormone (GRH)-stimulated GH release in sheep. Growth hormone secretory characteristics, the GH response to GRH, and steady-state GH mRNA concentrations were determined in castrated male lambs treated with 2 different doses of estradiol 17-beta for a 28-d experimental period. Although no differences between treatments in mean GH, basal GH, or GH pulse number were observed after 28 d of estradiol treatment, GH pulse amplitude was greater (P < 0.05) in the 2.00-cm implant-treated animals than in the control and 0.75-cm implant group. The effect of estradiol treatment on GRH-stimulated GH release revealed differences between the control and estradiol-treated animals (P < 0.05). The 15-min GH responses to 0.075 microg/kg hGRH in the control, 0.75-cm, and 2.00-cm implant groups, respectively, were 76 +/- 10, 22.6 +/- 2.1, and 43.6 +/- 15.0 ng/mL. Growth hormone mRNA content was determined for pituitary glands from the different treatment groups, and no differences in steady-state GH mRNA levels were observed. There were no differences in the mean plasma concentrations of IGF-I, cortisol, T(3), or T(4) from weekly samples. Growth hormone release from cultured ovine pituitary cells from control sheep was not affected by estradiol after 72 h or in a subsequent 3-h incubation with estradiol combined with GRH. These data suggest that estradiol has differing actions on basal and GRH-stimulated GH concentrations in plasma, but the increase in pulse amplitude does not represent an increased pituitary sensitivity to GRH.


Assuntos
Estradiol/farmacologia , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Hormônio do Crescimento/metabolismo , Ovinos/fisiologia , Animais , Northern Blotting/veterinária , Hormônio do Crescimento/genética , Hormônio do Crescimento/fisiologia , Hidrocortisona/sangue , Hidrocortisona/fisiologia , Immunoblotting/veterinária , Fator de Crescimento Insulin-Like I/fisiologia , Análise dos Mínimos Quadrados , Masculino , Hipófise/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Distribuição Aleatória , Tiroxina/sangue , Tiroxina/fisiologia , Tri-Iodotironina/sangue , Tri-Iodotironina/fisiologia
16.
Rev. chil. endocrinol. diabetes ; 2(3): 173-178, jul. 2009. ilus, tab
Artigo em Espanhol | LILACS | ID: lil-610296

RESUMO

Human growth is a complex process regulated by several genes, most of which are unknown. Recently, our knowledge regarding the etiology of genetically determined causes of short stature has greatly increased, so molecular analysis is becoming essential for the diagnosis of growth retardation. The advances in our understanding of the molecular mechanisms involved in the function of the somatotrophic axis have resulted in a dramatic enhancement of our ability to diagnose and treat growth disorders. We hope that in the next few years improved methods for identifying specific abnormalities which cause short stature will expand our ability to diagnose other causes of growth retardation, and reduce the proportion of patients with "idiopathic" short stature.


Assuntos
Humanos , Estatura/genética , Hormônio do Crescimento/genética , Transtornos do Crescimento/diagnóstico , Transtornos do Crescimento/genética , Fator de Crescimento Insulin-Like I/fisiologia , Fator de Crescimento Insulin-Like I/genética , Hipófise/fisiologia , Hipotálamo/fisiologia , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio do Crescimento/fisiologia , Mutação
17.
Am J Physiol Endocrinol Metab ; 296(5): E1085-92, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19240251

RESUMO

Age, sex steroids, and abdominal-visceral fat (AVF) jointly affect pulsatile growth hormone (GH) secretion. Pulsatile GH secretion in turn is controlled by GH-releasing hormone (GHRH), GH-releasing peptide (GHRP), and somatostatin. Marked stimulation of pulsatile GH secretion is achieved via GHRH-GHRP synergy. Nonetheless, how key modulators of GH secretion, such as age, sex steroids, and body mass index, modify GHRH-GHRP synergy is not known. The present strategy was to 1) infuse GHRH and GHRP-2 simultaneously to evoke synergy and 2) downregulate the gonadal axis with leuprolide and then restore placebo (Pl) or testosterone (T) to clamp the sex steroid milieu. Forty-seven men [18-74 yr of age, T = 7-1,950 ng/dl, estradiol (E(2)) = 5-79 pg/ml, insulin-like growth factor (IGF)-I = 115-817 microg/l, AVF = 11-349 cm(2)] were studied. GHRH-GHRP synergy correlated negatively with age and AVF (both P < 0.001) and positively with IGF-I (P < 0.001) and IGF-binding protein (IGFBP)-3 (P = 0.031). Unstimulated basal (nonpulsatile) GH secretion correlated positively with T (P = 0.015) and E(2) (P = 0.004) concentrations. Fasting pulsatile GH secretion varied negatively with age (P = 0.017) and positively with IGF-I (P = 0.002) and IGFBP-3 (P = 0.001). By stepwise forward-selection multivariate analyses, AVF, IGF-I, and IGFBP-3 together explained 60% of the variability in GHRH-GHRP synergy (P < 0.001), E(2) accounted for 17% of the variability in basal GH secretion (P = 0.007), and IGF-I explained 20% of the variability in fasting pulsatile GH secretion (P = 0.002). In conclusion, a paradigm examining GHRH-GHRP synergy under a sex steroid clamp reveals highly selective control of basal, pulsatile, and synergistic peptide-driven GH secretion by AVF, E(2), and IGF-I in healthy men.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/administração & dosagem , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Fator de Crescimento Insulin-Like I/metabolismo , Gordura Intra-Abdominal/fisiologia , Oligopeptídeos/administração & dosagem , Oligopeptídeos/fisiologia , Adolescente , Adulto , Fatores Etários , Idoso , Estudos de Coortes , Método Duplo-Cego , Estradiol/sangue , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/sangue , Gordura Intra-Abdominal/efeitos dos fármacos , Gordura Intra-Abdominal/metabolismo , Leuprolida/administração & dosagem , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Oligopeptídeos/metabolismo , Estudos Prospectivos , Análise de Regressão , Globulina de Ligação a Hormônio Sexual/metabolismo , Testosterona/administração & dosagem , Testosterona/sangue , Adulto Jovem
18.
Domest Anim Endocrinol ; 36(4): 186-96, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19157766

RESUMO

Corticosterone (CORT) can stimulate growth hormone (GH) secretion on embryonic day (e) 12 in the chicken. However, CORT failed to induce GH secretion on e20 in a single report, suggesting that regulation of GH production changes during embryonic development. Secretion in response to CORT during embryonic development is modulated by the thyroid hormones triiodothyronine (T(3)) and thyroxine (T(4)). Growth hormone responses on e12 involve both glucocorticoid (GR) and mineralocorticoid receptors (MR); however, involvement of MR has not been evaluated past e12. To further define changes in somatotroph responsiveness to CORT, pituitary cells obtained on e12-e20 were cultured with CORT alone and in combination with T(3) and GH-releasing hormone (GHRH). Growth hormone mRNA levels and protein secretion were quantified by quantitative real-time polymerase chain reaction (qRT-PCR) and radioimmunoassay (RIA), respectively. Corticosterone significantly increased GH mRNA and protein secretion on e12; however, mRNA concentration and protein secretion were unaffected on e20. Contributions of GR and MR in CORT responses were evaluated using GR and MR antagonists. Treatment with a GR-specific antagonist effectively blocked the CORT-induced increase in GH secretion on e12. The same treatment on e20 had no effect on GH secretion. These findings demonstrate that GR is directly involved in glucocorticoid stimulation of GH secretion at the time of somatotroph differentiation but is not regulatory at the end of embryonic development. We conclude that positive somatotroph responses to CORT are lost during chicken embryonic development and that GR is the primary regulator of CORT-induced GH secretion.


Assuntos
Corticosterona/fisiologia , Hormônio do Crescimento/metabolismo , Receptores de Glucocorticoides/metabolismo , Somatotrofos/metabolismo , Tri-Iodotironina/fisiologia , Animais , Células Cultivadas , Embrião de Galinha , Embrião não Mamífero , Desenvolvimento Embrionário/fisiologia , Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Adeno-Hipófise/citologia , Adeno-Hipófise/embriologia , RNA Mensageiro/análise
19.
Br J Cancer ; 98(11): 1790-6, 2008 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-18506184

RESUMO

Splice Variant 1 (SV-1) of growth hormone-releasing hormone (GHRH) receptor, found in a wide range of human cancers and established human cancer cell lines, is a functional receptor with ligand-dependent and independent activity. In the present study, we demonstrated by western blots the presence of the SV1 of GHRH receptor and the production of GHRH in MDA-MB-468, MDA-MB-435S and T47D human breast cancer cell lines, LNCaP prostate cancer cell line as well as in NCI H838 non-small cell lung carcinoma. We have also shown that GHRH produced in the conditioned media of these cell lines is biologically active. We then inhibited the intrinsic production of GHRH in these cancer cell lines using si-RNA, specially designed for human GHRH. The knocking down of the GHRH gene expression suppressed the proliferation of T47D, MDA-MB-435S, MDA-MB-468 breast cancer, LNCaP prostate cancer and NCI H838 non-SCLC cell lines in vitro. However, the replacement of the knocked down GHRH expression by exogenous GHRH (1-29)NH(2) re-established the proliferation of the silenced cancer cell lines. Furthermore, the proliferation rate of untransfected cancer cell lines could be stimulated by GHRH (1-29)NH(2) and inhibited by GHRH antagonists MZ-5-156, MZ-4-71 and JMR-132. These results extend previous findings on the critical function of GHRH in tumorigenesis and support the role of GHRH as a tumour growth factor.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/antagonistas & inibidores , Neoplasias/terapia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Hormônio Liberador de Hormônio do Crescimento/análise , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Humanos , Masculino , Neoplasias/patologia , RNA Interferente Pequeno/genética , Radioimunoensaio , Sermorelina/análogos & derivados , Sermorelina/farmacologia
20.
Exp Brain Res ; 188(4): 483-91, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18421448

RESUMO

In the rat, the sexual dimorphism in growth hormone release is driven by sex steroids, and is suggested to result mainly from differences in somatostatin (SOM) release patterns from the median eminence. We studied the effect of gonadal steroids on SOM peptide-containing cells in the periventricular nucleus (PeVN) of ovariectomized (OVX) female rats, and compared these data with data from intact male rats. Adult female rats were treated with estradiol (E(2)) and/or progesterone (P), 3 months (long-term) or 2 weeks (short-term) after ovariectomy (OVX). Perfusion-fixed brains were sliced and stained, and the number of SOM-immunoreactive (-ir) cells and total SOM-ir area (in microm(2)) were determined using computer assisted analysis. SOM-ir cells in the PeVN showed a very characteristic rostro-caudal distribution and localization in relation to the third ventricle. Both the number of SOM-ir cells and total SOM-ir area in the PeVN were higher in male compared to OVX female rats. Neither the number of SOM-ir cells, nor the total SOM-ir area in the PeVN was affected by E(2) or P treatment alone. Treatment with both gonadal steroids, however, did increase total SOM-immunoreactivity. This study is the first to describe SOM cell distribution within the rat PeVN in great detail. A clear sex difference exists in SOM peptide content in the rat PeVN. In addition, E(2) and P may act synergistically to affect SOM cells in the female PeVN, suggesting that both gonadal steroids may be involved in the generation of the typical feminine SOM release pattern.


Assuntos
Estradiol/farmacologia , Núcleos da Linha Média do Tálamo/metabolismo , Progesterona/farmacologia , Somatostatina/metabolismo , Animais , Feminino , Hormônio do Crescimento/metabolismo , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Imuno-Histoquímica , Masculino , Núcleos da Linha Média do Tálamo/efeitos dos fármacos , Ovariectomia , Perfusão , Ratos , Ratos Wistar , Caracteres Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA