Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-30114526

RESUMO

Phoenixin (Pnx) is an endogenous peptide known to be involved in reproduction and food intake in rats, with two active isoforms, phoenixin-14 (Pnx-14) and phoenixin-20 (Pnx-20). However, little is known about the functions of Pnx in teleost. Here, pnx was cloned and was detected in all tissues of both male and female in spotted scat (Scatophagus argus), including growth axis, hypothalamus, pituitary, and liver. Real-time PCR analysis showed that pnx in the hypothalamus increased significantly after 2 d and 7 d fasting, while reduced significantly after re-feeding (P < 0.05). When pituitary and liver fragments were cultured in vitro with Pnx-14 and Pnx-20 (10 nM and 100 nM) for 6 h, the expression of ghrhr (growth hormone-releasing hormone receptor) and gh (growth hormone) in the pituitary, and ghr1 (growth hormone receptor 1) in the liver increased significantly, except ghr2 (growth hormone receptor 2) incubated with 10 nM and 100 nM Pnx-20 and ghr1 incubated with 10 nM Pnx-20. Similarly, the expression of ghrhr and gh in the pituitary, as well as ghr1 and ghr2 in the liver, increased significantly after injecting S. argus with Pnx-14 and Pnx-20 (10 ng/g and 100 ng/g body weight). These results indicate that Pnx is likely to be involved in the regulation of food intake, and also regulates the growth of S. argus by increasing ghrhr and gh expression in the pituitary, ghr1 and ghr2 in the liver, and ghr1 directly in the liver.


Assuntos
Ingestão de Energia , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Hormônios Peptídicos/metabolismo , Perciformes/fisiologia , Animais , Aquicultura , China , Ingestão de Energia/efeitos dos fármacos , Feminino , Proteínas de Peixes/administração & dosagem , Proteínas de Peixes/genética , Proteínas de Peixes/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hormônio do Crescimento/agonistas , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Hormônios Hipotalâmicos/administração & dosagem , Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/farmacologia , Hipotálamo/efeitos dos fármacos , Injeções Intraperitoneais , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Especificidade de Órgãos , Hormônios Peptídicos/administração & dosagem , Hormônios Peptídicos/genética , Hormônios Peptídicos/farmacologia , Perciformes/crescimento & desenvolvimento , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Isoformas de Proteínas/administração & dosagem , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Distribuição Aleatória , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/agonistas , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Receptores da Somatotropina/agonistas , Receptores da Somatotropina/genética , Receptores da Somatotropina/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Técnicas de Cultura de Tecidos/veterinária , Aumento de Peso
2.
Growth Horm IGF Res ; 38: 14-18, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29277338

RESUMO

Several acquired or congenital hypothalamic abnormalities may cause growth failure (GF). We described two of these congenital abnormalities. First, a case of CHARGE syndrome, an epigenetic disorder mostly caused by heterozygous mutations in the gene encoding CHD7, a chromatin remodeling protein, causing several malformations, some life-threatening, with additional secondary hypothalamus-hypophyseal dysfunction, including GF. Second, a cohort of individuals with genetic isolated severe GH deficiency (IGHD), due to a homozygous mutation in the GH-releasing hormone (GHRH) receptor gene described in Itabaianinha County, in northeast Brazil. In this IGHD, with marked reduction of serum concentrations of IGF-I, and an up regulation of IGF-II, GF is the principal finding in otherwise normal subjects, with normal quality of life and longevity. This IGHD may unveil the effects of GHRH, pituitary GH and IGF-I, IGF-II and local GH and growth factor on the size and function of body and several systems. For instance, anterior pituitary hypoplasia, and impairment of the non-REM sleep may be due to GHRH resistance. Proportionate short stature, doll facies, high-pitched pre-pubertal voice, and reduced muscle mass reflect the lack of the synergistic effect of pituitary GH and IGF-I in bones and muscles. Central adiposity may be due to a direct effect of the lack of GH. Brain, eyes and immune system may also involve IGF-II and local GH or growth factors. A concept of physiological hierarchy controlling body size and function by each component of the GH system may be drawn from this model.


Assuntos
Síndrome CHARGE/etiologia , Nanismo Hipofisário/etiologia , Transtornos do Crescimento/etiologia , Hipotálamo/anormalidades , Mutação , Receptores de Neuropeptídeos/deficiência , Receptores de Hormônios Reguladores de Hormônio Hipofisário/deficiência , Adulto , Pré-Escolar , Feminino , Humanos , Masculino , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética
3.
Endocrinology ; 157(10): 3972-3982, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27253996

RESUMO

Isolated GH deficiency (IGHD) type II, the autosomal dominant form of GHD, is mainly caused by mutations that affect splicing of GH-1. When misspliced RNA is translated, it produces a toxic 17.5-kDa GH isoform that reduces the accumulation and secretion of wild-type-human GH (wt-hGH). Usually, isolated GHD type II patients are treated with daily injections of recombinant human GH in order to maintain normal growth. However, this type of replacement therapy does not prevent toxic effects of the 17.5-kDa GH isoform on the pituitary gland, which can eventually lead to other hormonal deficiencies. Here, we tested the possibility to restore the constitutive splicing pattern of GH-1 by using butyrate, a drug that mainly acts as histone deacetylase inhibitor. To this aim, wt-hGH and/or different hGH-splice site mutants (GH-IVS3+2, GH-IVS3+6, and GH-ISE+28) were transfected in rat pituitary cells expressing human GHRH receptor (GHRHR) (GC-GHRHR). Upon butyrate treatment, GC-GHRHR cells coexpressing wt-hGH and each of the mutants displayed increased GH transcript level, intracellular GH content, and GH secretion when compared with the corresponding untreated condition. The effect of butyrate was most likely mediated by the alternative splicing factor/splicing factor 2. Overexpression of alternative ASF/SF2 in the same experimental setting, indeed, promoted the amount of full-length transcripts thus increasing synthesis and secretion of the 22-kDa GH isoform. In conclusion, our results support the hypothesis that modulation of GH-1 splicing pattern to increase the 22-kDa GH isoform levels can be clinically beneficial and hence a crucial challenge in GHD research.


Assuntos
Butiratos/uso terapêutico , Nanismo Hipofisário/tratamento farmacológico , Hormônio do Crescimento/metabolismo , Splicing de RNA/efeitos dos fármacos , Animais , Butiratos/farmacologia , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Técnicas de Transferência de Genes , Hormônio do Crescimento/genética , Humanos , Ratos , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo
4.
Endocr J ; 61(1): 25-34, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24088662

RESUMO

Gonadotropin-inhibitory hormone (GnIH) was first identified in quail as a novel neurohormone that acts directly on the anterior pituitary to inhibit gonadotropin release. GnIH inhibits not only gonadotropin release from the pituitary gland but also inhibits the release of gonadotropin-releasing hormone (GnRH) from the hypothalamus. In this study, we examined how GnIH receptors were regulated in pituitary gonadotroph cells and GnRH-producing neurons in the hypothalamus. In the mouse pituitary gonadotroph cell line LßT2, GnRH increased expression of the GnIH receptor, G-protein coupled receptor 74 (GPR74). GnRH also stimulated the expression of GPR74 and GPR147 in primary cultures of rat anterior pituitary cells. In addition, when GnRH was administered to LßT2 cells in a pulsatile manner, low frequency GnRH pulse stimulation stimulated GPR74 and GPR147 expression more than did high frequency GnRH pulses. In the mouse hypothalamic GnRH-producing cell line GT1-7, hypothalamic kisspeptin did not significantly increase the expression of GnIH receptors. However, the intermittent administration of kisspeptin to GT1-7 cells significantly increased GPR74 and GPR147 mRNA expression. The overexpression of either constitutively active MEK kinase (MEKK) or protein kinase A (PKA) in LßT2 cells increased the expression of GPR74 mRNA. Conversely, in GT1-7 cells, although the overexpression of either MEKK or PKA failed to stimulate GnIH receptor expression, the combined overexpression of both kinases together increased GPR74 and GPR147 mRNA levels. Our current observations suggest that two central controllers of reproductive function, GnRH and kisspeptin, stimulate the expression of GnIH receptors in pituitary gonadotroph cells and hypothalamic GnRH neurons.


Assuntos
Expressão Gênica , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/biossíntese , Hipotálamo/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Animais , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/genética , Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/administração & dosagem , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Kisspeptinas/farmacologia , MAP Quinase Quinase Quinases/genética , Camundongos , RNA Mensageiro/análise , Ratos , Receptores de Neuropeptídeos/genética
5.
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
6.
Reprod Biol Endocrinol ; 8: 54, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20509930

RESUMO

BACKGROUND: Antagonists of growth hormone-releasing hormone (GHRH) are being developed for the treatment of various human cancers. METHODS: MTT assay was used to test the proliferation of SKOV3 and CaOV3. The splice variant expression of GHRH receptors was examined by RT-PCR. The expression of protein in signal pathway was examined by Western blotting. siRNA was used to block the effect of EGFR. RESULTS: In this study, we investigated the effects of a new GHRH antagonist JMR-132, in ovarian cancer cell lines SKOV3 and CaOV3 expressing splice variant (SV)1 of GHRH receptors. MTT assay showed that JMR-132 had strong antiproliferative effects on SKOV3 and CaOV3 cells in both a time-dependent and dose-dependent fashion. JMR-132 also induced the activation and increased cleaved caspase3 in a time- and dose-dependent manner in both cell lines. In addition, JMR-132 treatments decreased significantly the epidermal growth factor receptor (EGFR) level and the phosphorylation of Akt (p-Akt), suggesting that JMR-132 inhibits the EGFR-Akt pathway in ovarian cancer cells. More importantly, treatment of SKOV3 and CaOV3 cells with 100 nM JMR-132 attenuated proliferation and the antiapoptotic effect induced by EGF in both cell lines. After the knockdown of the expression of EGFR by siRNA, the antiproliferative effect of JMR-132 was abolished in SKOV3 and CaOV3 cells. CONCLUSIONS: The present study demonstrates that the inhibitory effect of the GHRH antagonist JMR-132 on proliferation is due, in part, to an interference with the EGFR-Akt pathway in ovarian cancer cells.


Assuntos
Adenocarcinoma/patologia , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/fisiologia , Neoplasias Ovarianas/patologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Sermorelina/análogos & derivados , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antineoplásicos Hormonais/farmacologia , Avaliação Pré-Clínica de Medicamentos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Hormônio do Crescimento/antagonistas & inibidores , Antagonistas de Hormônios/farmacologia , Humanos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/farmacologia , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Sermorelina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas
7.
Fertil Steril ; 94(3): 841-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19524226

RESUMO

OBJECTIVE: To determine the effect of the GHRH antagonist JV-1-36 on proliferation and survival of primary ectopic human endometriotic stromal cells (ESCs) and the T HESC cell line. DESIGN: Prospective laboratory study. SETTING: University hospital. PATIENT(S): 22 women with endometriosis (aged 34.8+/-5.7 years) undergoing therapeutic laparoscopy. INTERVENTION(S): Eutopic (n=10) and ectopic (n=22) endometrial tissues were collected from women who underwent therapeutic laparoscopic surgery for endometriosis (stage III/IV). MAIN OUTCOME MEASURE(S): Expression of GHRH, GHRH receptor (GHRH-R) and GHRH-R splice variant (SV) 1 mRNA was determined by reverse-transcription polymerase chain reaction (RT-PCR). The ESC proliferation was assessed by 5-bromo-2-deoxyuridine incorporation, cell survival by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) and Trypan blue assay. The T HESC survival was evaluated by MTT, cyclic adenosine monophosphate (cAMP) levels by ELISA, extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylation by Western blot, and insulin-like growth factor (IGF)-2 mRNA by real-time PCR. RESULT(S): The ESCs and T HESCs, but not normal endometrial tissues, expressed GHRH-R mRNA; SV1 mRNA was determined in normal endometrial tissues, ESCs, and T HESCs; GHRH mRNAwas found in T HESCs; JV-1-36 inhibited ESC proliferation and ESC and T HESC survival. In T HESCs, JV-1-36 reduced cAMP production and ERK1/2 phosphorylation but had no effect on IGF-2 mRNA expression. CONCLUSION(S): The GHRH antagonist JV-1-36 inhibits endometriotic cell proliferation and survival, suggesting that GHRH antagonist may represent promising tools for treatment of endometriosis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Coristoma/patologia , Endometriose/patologia , Hormônio Liberador de Hormônio do Crescimento/análogos & derivados , Células Estromais/efeitos dos fármacos , Doenças Uterinas/patologia , Adulto , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Coristoma/genética , Coristoma/metabolismo , Avaliação Pré-Clínica de Medicamentos , Endometriose/genética , Endometriose/metabolismo , Endométrio , Feminino , Hormônio Liberador de Hormônio do Crescimento/antagonistas & inibidores , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Antagonistas de Hormônios/farmacologia , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Células Estromais/fisiologia , Doenças Uterinas/genética , Doenças Uterinas/metabolismo
8.
Int J Oncol ; 32(3): 593-601, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18292936

RESUMO

The expression of growth hormone-releasing hormone (GHRH) and its receptors has been demonstrated in peripheral tissues as well as CNS. Recently, the functional splice variant SV1 of GHRH receptor was identified in various human cancers and cancer cell lines. Although antineoplastic activity of GHRH antagonists has been clearly demonstrated, the mechanism of action is incompletely understood. The objective of this study was the investigation of direct anti-proliferative effect of GHRH antagonist MZ-5-156 on HEC-1A human endometrial cancer cell line and the elucidation of underlying mechanisms. RT-PCR revealed the expression of mRNA for GHRH and SV1 of GHRH receptor in HEC-1A cells. MZ-5-156, at concentrations between 10(-7) and 10(-5) M, had a dose-dependent antiproliferative effect on HEC-1A cells, as determined by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, (MTS) assay. Hoechst 33342 staining and flow cytometric analysis indicated that MZ-5-156, at 10(-6) M, induced apoptosis in HEC-1A cells after 48 h of treatment. Western blot analysis of apoptosis-related proteins demonstrated that treatment with MZ-5-156 (10(-6) M) for 48 h significantly increased the protein levels of Fas, phospho-p53 (Ser46), p53AIP1 (p53-regulated Apoptosis-Inducing Protein 1), and caspase-8, -9, and -3, and decreased the protein level of Bcl-2. These results demonstrate that MZ-5-156 can directly inhibit the proliferation of human endometrial cancer cells, which express mRNA for GHRH and SV1 of GHRH receptor, presumably through the induction of p53-dependent apoptosis coupled with the up-regulation of Fas, phospho-p53 (Ser46), p53AIP1, and caspase-8, -9, and -3, and the down-regulation of Bcl-2.


Assuntos
Adenocarcinoma Papilar/patologia , Proliferação de Células/efeitos dos fármacos , Neoplasias do Endométrio/patologia , Hormônio Liberador de Hormônio do Crescimento/antagonistas & inibidores , Sermorelina/análogos & derivados , Adenocarcinoma Papilar/genética , Idoso , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Caspases/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Neoplasias do Endométrio/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Genes bcl-2 , Genes p53 , Hormônio Liberador de Hormônio do Crescimento/genética , Humanos , RNA Mensageiro/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Sermorelina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
9.
J Endocrinol ; 195(3): 525-36, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18000314

RESUMO

In this study, two novel GHRHR receptor splice variants, named chicken GHRHR-v1 (cGHRHR-v1) and cGHRHR-v2 respectively, were identified from chicken pituitary using RT-PCR assay. cGHRHR-v1 is characterized by an N-terminal deletion of 36 amino acid residues, including an aspartate at position 56 (Asp(56)) conserved in G protein-coupled receptor B-I subfamily. cGHRHR-v2 is a carboxyl-terminal truncated receptor variant with four putative transmembrane domains, which arose from alternative use of a splice acceptor site on intron 8. Using the pGL3-CRE-luciferase reporter system, the functionality of the two variants was examined in Chinese hamster ovary cells. cGHRHR-v1 was shown to be capable of transmitting signal upon agonist stimulation, but cGHRHR-v2 could not. Both GHRH and pituitary adenylate cyclase-activating peptide (PACAP) could activate cGHRHR-v1 at high dosages (GHRH >/=10(-8) M; PACAP >/=10(-6) M) and GHRH was much more potent than PACAP, suggesting that cGHRHR-v1 is a functional membrane-spanning receptor with an impairment in high-affinity ligand binding, rather than in receptor activation and ligand-binding specificity. This finding also points out the possibility that Asp(56) is not a critical determinant for receptor activation and direct ligand-receptor interaction. To substantiate this hypothesis, using site-directed mutagenesis, two receptor mutants with replacement of Asp(56) by Ala or Gly were generated. Expectedly, chicken or human GHRH could still activate both receptor mutants with reduced potencies (about 2- to 14-fold less potent). Taken together, our findings not only suggest that cGHRHR variants may play a role in controlling normal pituitary functions, but also support that Asp(56) is nonessential for receptor activation and direct ligand-receptor interaction.


Assuntos
Galinhas/genética , Galinhas/metabolismo , Isoformas de Proteínas , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Sequência de Aminoácidos , Animais , Ácido Aspártico , Sequência de Bases , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Humanos , Ligantes , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Hipófise/metabolismo , Receptores de Neuropeptídeos/agonistas , Receptores de Neuropeptídeos/efeitos dos fármacos , Receptores de Hormônios Reguladores de Hormônio Hipofisário/agonistas , Receptores de Hormônios Reguladores de Hormônio Hipofisário/efeitos dos fármacos , Ovinos , Transdução de Sinais
10.
Am J Physiol Regul Integr Comp Physiol ; 293(2): R922-30, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17537840

RESUMO

Growth hormone-releasing hormone (GHRH), its receptor (GHRHR), and other members of the somatotropic axis are involved in non-rapid eye movement sleep (NREMS) regulation. Previously, studies established the involvement of hypothalamic GHRHergic mechanisms in NREMS regulation, but cerebral cortical GHRH mechanisms in sleep regulation remained uninvestigated. Here, we show that unilateral application of low doses of GHRH to the surface of the rat somatosensory cortex ipsilaterally decreased EEG delta wave power, while higher doses enhanced delta power. These actions of GHRH on EEG delta wave power occurred during NREMS but not during rapid eye movement sleep. Further, the cortical forms of GHRH and GHRHR were identical to those found in the hypothalamus and pituitary, respectively. Cortical GHRHR mRNA and protein levels did not vary across the day-night cycle, whereas cortical GHRH mRNA increased with sleep deprivation. These results suggest that cortical GHRH and GHRHR have a role in the regulation of localized EEG delta power that is state dependent, as well as in their more classic hypothalamic role in NREMS regulation.


Assuntos
Ritmo Delta , Hormônio Liberador de Hormônio do Crescimento/fisiologia , Receptores de Neuropeptídeos/fisiologia , Receptores de Hormônios Reguladores de Hormônio Hipofisário/fisiologia , Fases do Sono/fisiologia , Córtex Somatossensorial/fisiologia , Animais , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Hipotálamo/fisiologia , Masculino , Microinjeções , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Privação do Sono/fisiopatologia , Fases do Sono/efeitos dos fármacos , Córtex Somatossensorial/efeitos dos fármacos
11.
Endocrinology ; 147(5): 2535-43, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16469800

RESUMO

GHRH stimulates GH secretion in chickens as in mammals. However, nothing is known about the chicken GHRH receptor (GHRH-R). Here we report the cDNA sequence of chicken GHRH-R. Comparison of the cDNA sequence with the chicken genome localized the GHRH-R gene to chicken chromosome 2 and indicated that the chicken GHRH-R gene consists of 13 exons. Expression of all exons was confirmed by RT-PCR amplification of pituitary mRNA. The amino acid sequence predicted by the GHRH-R cDNA is homologous to that in other vertebrates and contains seven transmembrane domains and a conserved hormone-binding domain. The predicted size of the GHRH-R protein (48.9 kDa) was confirmed by binding of (125)I-GHRH to chicken pituitary membranes and SDS-PAGE. GHRH-R mRNA was readily detected by RT-PCR in the pituitary but not in the hypothalamus, total brain, lung, adrenal, ovary, or pineal gland. Effects of corticosterone (CORT), GHRH, ghrelin, pituitary adenylate cyclase-activating peptide, somatostatin (SRIF), and TRH on GHRH-R and GH gene expression were determined in cultures of chicken anterior pituitary cells. GHRH-R and GH mRNA levels were determined by quantitative real-time RT-PCR. Whereas all treatments affected levels of GH mRNA, only CORT, GHRH, and SRIF significantly altered GHRH-R mRNA levels. GHRH-R gene expression was modestly increased by GHRH and suppressed by SRIF at 4 h, and CORT dramatically decreased levels of GHRH-R mRNA at 72 h. We conclude that adrenal glucocorticoids may substantially impact pituitary GH responses to GHRH in the chicken through modulation of GHRH-R gene expression.


Assuntos
Regulação da Expressão Gênica , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/fisiologia , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/fisiologia , Sequência de Aminoácidos , Animais , Encéfalo/embriologia , Células Cultivadas , Galinhas , Mapeamento Cromossômico , Biologia Computacional , Reagentes de Ligações Cruzadas/farmacologia , Primers do DNA/química , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Éxons , Feminino , Biblioteca Gênica , Hipotálamo/metabolismo , Íntrons , Dados de Sequência Molecular , Peso Molecular , Filogenia , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Distribuição Tecidual
12.
Vitam Horm ; 70: 1-24, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15727800

RESUMO

Growth hormone-releasing hormone (GHRH) is a neuropeptide secreted by the hypothalamus that stimulates the synthesis and release of growth hormone (GH) in the pituitary. Accumulating evidence suggests that in addition to GHRH's neuroendocrine action, GHRH is present in several extrahypothalamic tissues and is involved in a variety of cellular processes. Its function is related to the regulation of cell proliferation and differentiation of various nonpituitary cell types. In certain cases, ectopic production of GHRH has also been implicated in carcinogenesis. The mechanisms by which GHRH affects the peripheral extrapituitary tissues remain poorly understood, but it is likely that classic neuroendocrine action as well as paracrine and autocrine pathways are involved. Some headway has been made in the identification of extrapituitary receptors for GHRH and cDNA as splice variants of these GHRH receptors found in various tumors. The fact that the nonpituitary GHRH receptors are not fully identified, however, remains the major obstacle in studying, at a more mechanistic level, the action of local GHRH. This review summarizes the information available regarding the role of GHRH in the extrapituitary tissues with emphasis on its potential therapeutic and diagnostic applications.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/fisiologia , Hipófise/fisiologia , Animais , Diferenciação Celular , Divisão Celular , Hormônios Ectópicos , Humanos , Hipotálamo/fisiologia , Neoplasias , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/fisiologia , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/fisiologia
13.
Exp Gerontol ; 39(11-12): 1761-4, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15582293

RESUMO

We briefly compare calorie restriction, GHRH-R and Pit-1 mutants with knockout phenotypes of GH receptor, IGF-1 receptor and p66Shc, to make some general conclusions. Growth, fertility and longevity phenotypes may dissociate in some of these mutants, and we try to interpret this. Follows a short discussion on the importance of genetic background for aging studies in mice. We then evoke studies in C. elegans showing that lifespan may be regulated in a non-cell-autonomous fashion, and that the nervous system could play a central role therein. Recent findings on DILP-2 regulation in Drosophila transpose this hypothesis of endocrine lifespan regulation to insects. Work in mice shows that inactivation of the insulin receptor specifically in the adipose tissue is sufficient to increase the mouse lifespan. In summary, exciting findings obtained in very different model organisms are rapidly converging and suggest that animal lifespan may be subject to endocrine regulation. Interestingly, the hypothalamus centralizes many age related hormonal regulations and at the same time participates in the integration of numerous nutritional signals, such that one could ask whether the hypothalamus may be at the crossroads of metabolic and endocrine lifespan regulation.


Assuntos
Envelhecimento/fisiologia , Hipotálamo/fisiologia , Fator de Crescimento Insulin-Like I/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Privação de Alimentos , Hormônio do Crescimento/metabolismo , Longevidade , Masculino , Camundongos , Modelos Animais , Mutação , Hipófise/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Fator de Transcrição Pit-1 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
14.
Endocrinology ; 145(9): 4134-43, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15155578

RESUMO

The proliferation of pituitary somatotroph cells and the synthesis and secretion of GH are under the stimulatory control of the hypothalamic peptide GHRH. GHRH is initially synthesized as pre-prohormone and then enzymatically cleaved to its mature form (44 amino acids in humans and 42 in mice). Although mutations in the GHRH receptor cause isolated GH deficiency (IGHD) both in humans and mice, mutations in the GHRH gene have never been described. To determine the consequences of generalized lack of GHRH, we have created a mouse with targeted disruption (knockout) of the GHRH gene (GHRHKO). We have substituted a portion of the gene that encodes for the initial 14 amino acids of the 1-42 GHRH with a neomycin resistance cassette. Heterozygous founder (+/-) mice were mated to obtain -/- animals. The expected Mendelian ratio was conserved (25.8% of offspring were +/+, 52.8% were +/-, and 21.4% were -/-), showing no lethality in the GHRHKO embryos. GHRHKO mice appeared normal at birth. Starting at 3 wk of age, -/- mice showed significant growth retardation. By 12 wk of age, their weight was about 60% of +/+ and +/- littermates. Growth retardation was due to IGHD, as shown by reduced pituitary GH mRNA and protein content, reduced serum IGF-I, and reduced liver IGF-I mRNA. The phenotype of the GHRHKO mice is similar to the one observed in the mouse with mutated GHRH receptor, including pituitary hypoplasia. Heterozygous mice had normal growth, although adult +/- males (but not females) had mild reduction in serum IGF-I. In conclusion, we demonstrate that ablation of the GHRH gene causes IGHD in mice. The GHRHKO mouse will be the new useful model of IGHD.


Assuntos
Modelos Animais de Doenças , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio do Crescimento/deficiência , Camundongos Knockout , Animais , Fertilidade , Hormônio do Crescimento/metabolismo , Hipotálamo/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Fígado/fisiologia , Camundongos , Hipófise/metabolismo , Hipófise/patologia , Prolactina/metabolismo , RNA Mensageiro/análise , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética
15.
J Neuroendocrinol ; 16(2): 113-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14763997

RESUMO

Changes in growth hormone-releasing hormone (GHRH), GHRH-receptor (R), somatostatin and interleukin (IL)-1beta mRNA levels were determined in fetal rat hypothalamic cultures after administration of IL-1beta (1, 10, 100 ng/ml, 2 h incubation), and in adult rat hypothalamus 5 h after intracerebroventricular injection of IL-1beta (2.5 and 25 ng). IL-1beta stimulated GHRH-R mRNA expression both in vitro (10 and 100 ng/ml) and in vivo (2.5 and 25 ng). Somatostatin mRNA was significantly stimulated and GHRH mRNA slightly reduced in vitro, while these mRNA species were not altered in vivo in response to IL-1beta. IL-1beta stimulated its own expression both in vitro (10 and 100 ng/ml) and in vivo (25 ng). IL-1beta-induced mRNA responses occurred 2 h after treatment in vitro (incubation times, 30 min to 6 h). IL-1beta also elicited slight GHRH releases in vitro. Up-regulation of hypothalamic GHRH-R by IL-1beta may explain previous findings suggesting that IL-1beta stimulates GHRH activity.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hipotálamo/metabolismo , Interleucina-1/fisiologia , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Somatostatina/metabolismo , Animais , Células Cultivadas , Feto , Hormônio Liberador de Hormônio do Crescimento/genética , Hipotálamo/citologia , Masculino , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Somatostatina/genética , Regulação para Cima
16.
DNA Seq ; 14(2): 129-33, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12825354

RESUMO

The genomic organisation of a putative GHRH receptor gene has been characterised in the teleost fish, Fugu rubripes. It comprises 13 exons and is at least 1.2 x larger than previously described vertebrate GHRH receptors. Sequence conservation with other vertebrate GHRH receptors is highest over the seven transmembrane domains with preservation of 8 conserved cysteines and an N-glycosylation site in the N-terminal region, which are important for receptor activation. Database mining revealed a further putative GHRH receptor in Fugu, with phylogenetic tree topology indicating that this was potentially a teleost-specific duplication event. This is the first time that the genomic organisation of a GHRH receptor gene has been characterised from a nonmammalian vertebrate.


Assuntos
Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Takifugu/genética , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , Cosmídeos , Cisteína/metabolismo , DNA Complementar/metabolismo , Éxons , Biblioteca Gênica , Ligação Genética , Glicosilação , Modelos Genéticos , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
17.
Am J Physiol Regul Integr Comp Physiol ; 284(1): R131-9, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12388430

RESUMO

The role of the somatotropic axis in sleep regulation was studied by using the lit/lit mouse with nonfunctional growth hormone (GH)-releasing hormone (GHRH) receptors (GHRH-Rs) and control heterozygous C57BL/6J mice, which have a normal phenotype. During the light period, the lit/lit mice displayed significantly less spontaneous rapid eye movement sleep (REMS) and non-REMS (NREMS) than the controls. Intraperitoneal injection of GHRH (50 microg/kg) failed to promote sleep in the lit/lit mice, whereas it enhanced NREMS in the heterozygous mice. Subcutaneous infusion of GH replacement stimulated weight gain, increased the concentration of plasma insulin-like growth factor-1 (IGF-1), and normalized REMS, but failed to restore normal NREMS in the lit/lit mice. The NREMS response to a 4-h sleep deprivation was attenuated in the lit/lit mice. In control mice, intraperitoneal injection of ghrelin (400 microg/kg) elicited GH secretion and promoted NREMS, and intraperitoneal administration of the somatostatin analog octretotide (Oct, 200 microg/kg) inhibited sleep. In contrast, these responses were missing in the lit/lit mice. The results suggest that GH promotes REMS whereas GHRH stimulates NREMS via central GHRH-Rs and that GHRH is involved in the mediation of the sleep effects of ghrelin and somatostatin.


Assuntos
Deleção de Genes , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Sono/fisiologia , Animais , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Grelina , Hormônio do Crescimento/administração & dosagem , Hormônio do Crescimento/farmacologia , Hormônio Liberador de Hormônio do Crescimento/administração & dosagem , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Octreotida/administração & dosagem , Octreotida/farmacologia , Hormônios Peptídicos/administração & dosagem , Hormônios Peptídicos/farmacologia , Fenótipo , Receptores de Neuropeptídeos/fisiologia , Receptores de Hormônios Reguladores de Hormônio Hipofisário/fisiologia , Sono/efeitos dos fármacos , Sono/genética , Sono REM/efeitos dos fármacos , Sono REM/genética , Sono REM/fisiologia
18.
J Clin Endocrinol Metab ; 87(11): 5076-84, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12414875

RESUMO

Pituitary stalk interruption and ectopic posterior lobe on magnetic resonance imaging (MRI) are frequently observed in patients with GH deficiency (GHD), but their pathogenesis remains controversial. We performed pituitary stimulation tests, MRI, and studied GH-1, GHRH receptor (GHRH-R), and Prophet of Pit-1 (PROP-1) genes in 76 patients with GHD. Of 33 patients with isolated GHD, 4 had GH-1 deletions and 4 had GHRH-R mutations; of 43 patients with combined pituitary hormone deficiency, 1 had PIT-1 and 5 had PROP-1 mutations. Compared with the 62 patients without mutations, 14 patients with mutations had higher frequency of consanguinity (57 vs. 2%, P < 0.001), familial cases (21 vs. 3%, P < 0.05), and lower frequency of breech delivery or hypoxemia at birth (0 vs. 39%, P < 0.005). On MRI, all patients with mutations had an intact stalk, whereas it was interrupted or thin in 74% without mutations (P < 0.001). The posterior pituitary lobe was in normal position in 92% of patients with mutations against 13% without mutations (P < 0.001). Among patients with combined pituitary hormone deficiency, hormonal deficiencies were of pituitary origin in all with PROP-1 and PIT-1 mutations and suggestive of hypothalamic origin in 81% without mutations. Perinatal insults were associated with thin/interrupted pituitary stalk, ectopic posterior lobe, and hypothalamic origin of hormonal deficiencies. In contrast, GH-1, GHRH-R, and PROP-1 mutations were associated with consanguineous parents, intact pituitary stalk, normal posterior lobe, and pituitary origin of hormonal deficiencies. We conclude that pituitary MRI and hormonal response to stimulation tests are useful in selection of patients and candidate genes to elucidate the etiological diagnosis of GHD.


Assuntos
Proteínas de Homeodomínio/genética , Hormônio do Crescimento Humano/deficiência , Hormônio do Crescimento Humano/genética , Mutação , Hipófise/patologia , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Adolescente , Adulto , Criança , Pré-Escolar , Consanguinidade , Feminino , Deleção de Genes , Humanos , Hipotálamo/fisiopatologia , Lactente , Imageamento por Ressonância Magnética , Masculino , Hipófise/fisiopatologia , Neuro-Hipófise/patologia
19.
J Neurosci ; 21(8): 2912-8, 2001 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-11306643

RESUMO

The somatotropic axis, and particularly growth hormone-releasing hormone (GHRH), is implicated in the regulation of sleep-wake activity. To evaluate sleep in chronic somatotropic deficiency, sleep-wake activity was studied in dwarf (dw/dw) rats that are known to have a defective GHRH signaling mechanism in the pituitary and in normal Lewis rats, the parental strain of the dw/dw rats. In addition, expression of GHRH receptor (GHRH-R) mRNA in the hypothalamus/preoptic region and in the pituitary was also determined by means of reverse transcription-PCR, and GHRH content of the hypothalamus was measured. Hypothalamic/preoptic and pituitary GHRH-R mRNA levels were decreased in the dw/dw rats, indicating deficits in the central GHRHergic transmission. Hypothalamic GHRH content in dw/dw rats was also less than that found in Lewis rats. The dw/dw rats had less spontaneous nonrapid eye movement sleep (NREMS) (light and dark period) and rapid eye movement sleep (REMS) (light period) than did the control Lewis rats. After 4 hr of sleep deprivation, rebound increases in NREMS and REMS were normal in the dw/dw rat. As determined by fast Fourier analysis of the electroencephalogram (EEG), the sleep deprivation-induced enhancements in EEG slow-wave activity in the dw/dw rats were only one-half of the response in the Lewis rats. The results are compared with sleep findings previously obtained in GHRH-deficient transgenic mice. The alterations in NREMS are attributed to the defect in GHRH signaling, whereas the decreases in REMS might result from the growth hormone deficiency in the dw/dw rat.


Assuntos
Nanismo Hipofisário/metabolismo , Hormônio Liberador de Hormônio do Crescimento/deficiência , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo , Transdução de Sinais , Transtornos Intrínsecos do Sono/metabolismo , Animais , Ritmo Circadiano , Nanismo Hipofisário/complicações , Nanismo Hipofisário/genética , Eletroencefalografia , Hormônio do Crescimento/deficiência , Hormônio Liberador de Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hipotálamo/metabolismo , Masculino , Hipófise/metabolismo , Área Pré-Óptica/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos Lew , Ratos Mutantes , Receptores de Neuropeptídeos/deficiência , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/deficiência , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Privação do Sono , Transtornos Intrínsecos do Sono/complicações , Transtornos Intrínsecos do Sono/diagnóstico , Fases do Sono/genética
20.
J Neuroendocrinol ; 11(4): 299-306, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10223284

RESUMO

Experimental evidence suggests that differential pituitary sensitivity to hypothalamic signals exerts a role in mediating both age and sex dependent patterns of growth hormone (GH) release and synthesis. One mechanism by which pituitary sensitivity to hypothalamic GH regulators could be modified is by the differential synthesis of their pituitary receptors. In the present report we therefore studied the age and sex dependency of the expression of receptors for two known stimulators of GH release, growth hormone-releasing hormone (GHRH) and the synthetic peptidyl and non-peptidyl GH secretagogues (GHSs). Pituitary GHRH receptor (GHRH-R) and GHS receptor (GHS-R) mRNA levels were measured by reverse transcriptase-polymerase chain reaction (RT-PCR) in male and female rats at postnatal day 1, 10, 30 and 75. We also examined the age- and sex-dependent expression of the GHS-R in whole hypothalamic extracts, since the GHS-R is also expressed in a variety of nuclei within the hypothalamus and has been linked to central regulation of the GH-axis. Pituitary GHRH-R mRNA concentrations were age-dependent; the highest levels were observed in d1 pituitaries and then declined with age, reaching a nadir by d30. These results are in concordance with the age-related decline in pituitary GHRH sensitivity. In contrast, the ontogenic pattern of GHS-R expression was bimodal; GHS-R mRNA concentrations in dl and d30 pituitaries were approximately twice those at d10 and d75. These results mirror the transient increase in GHS sensitivity observed around the onset of puberty, suggesting that gonadal steroids mediate GHS-R expression. GHRH-R mRNA levels were comparable in males and females within each age while GHS-R mRNA levels were gender dependent. At d30, male GHS-R mRNA levels were 30% greater than in their female counterparts. This was reversed at d75, when females had 89% more GHS-R mRNA per pituitary and 65% more per somatotrope than did age-matched males. These sexual differences further support a role for gonadal steroids in the modulation of pituitary GHS-R synthesis. The ontogenic and gender-specific pattern of hypothalamic GHS-R expression differed from that observed for the pituitary. Hypothalamic GHS-R mRNA levels increased with age but exhibited no significant sex difference at each age tested. Taken together, these data demonstrate that changes in the levels of pituitary GHS-R mRNA, but not GHRH-R mRNA, are associated with changes in the gonadal steroid environment, thereby implicating the GHS/GHS-R signalling system as a control point in the establishment and maintenance of sexually dimorphic patterns of GH secretion.


Assuntos
Hipotálamo/metabolismo , Hipófise/metabolismo , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Animais , Feminino , Hipotálamo/crescimento & desenvolvimento , Masculino , Hipófise/crescimento & desenvolvimento , Gravidez , RNA Mensageiro/genética , Ratos , Receptores de Grelina , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA