Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
J Neuroendocrinol ; 23(3): 197-207, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21166728

RESUMO

We have developed a system to use secreted fluorescent proteins (FPs) as surrogate markers for the continuous on-line monitoring of hormone release from perfused tissue slices. We have tested this system using GH-GFP transgenic rats with green fluorescent protein (GFP) targeted to the secretory vesicles (SVs) of pituitary growth hormone (GH) cells. Brief exposures of vibratome slices to GH secretagogues [GH-releasing hormone (GHRH), GH-releasing peptide-6 (GHRP-6)] or somatostatin caused changes in FP output that correlate with hormone secretion, subsequently measured in fractions of perfusate by radioimmunoassay. The temporal resolution of this method was capable of revealing differences in the kinetics of response to GHRH and GHRP-6 between wild-type and dwarf (dw/dw) rats harbouring the GH-GFP transgene. We further tested the utility of the system by generating transgenic mice with red FPs targeted to secretory vesicles (PRL-mRFP(sv)) and to the cytoplasm (PRL-DsRed(cyto)) of lactotrophs. Dopamine had no effect on the FP output from pituitary slices of PRL-DsRed(cyto) mice but inhibited output from those of PRL-mRFP(sv) animals, with a rebound increase of release after removal, which again correlated with hormone output measured in the perfusate by radioimmunoassay. The inhibition of monomeric RFP secretion by dopamine was dose-dependent, as was stimulation by low concentrations of oxytocin. The temporal resolution afforded by this method provides useful insight into the release kinetics from large populations of pituitary cells, and fills a temporo-spatial gap between single vesicle and single cell monitoring of exocytosis in milliseconds, and in vivo sampling studies of release into the bloodstream on a time scale of minutes.


Assuntos
Rastreamento de Células/métodos , Monitoramento de Medicamentos/métodos , Proteínas de Fluorescência Verde/metabolismo , Somatotrofos/metabolismo , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Rastreamento de Células/instrumentação , Monitoramento de Medicamentos/instrumentação , Nanismo/metabolismo , Nanismo/patologia , Células Endócrinas/efeitos dos fármacos , Células Endócrinas/metabolismo , Células Endócrinas/patologia , Feminino , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/farmacocinética , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Oligopeptídeos/farmacologia , Sistemas On-Line , Perfusão , Ratos , Ratos Transgênicos , Somatostatina/farmacologia , Somatotrofos/efeitos dos fármacos , Somatotrofos/patologia
2.
J Neuroendocrinol ; 20(11): 1278-87, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18752655

RESUMO

The ultradian pulsatile pattern of growth hormone (GH) secretion is markedly sexually dimorphic in rodents as in primates, but the neuroanatomical mechanisms of this phenomenon are not clear. In the arcuate nucleus of the hypothalamus, GH-releasing hormone (GHRH) neurones receive somatostatinergic inputs through the sst2A receptor (sst2A-R) and the percentage of GHRH neurones bearing sst2A-R is higher in female than in male GHRH-enhanced green fluorescent protein (eGFP) mice. In the present study, we hypothesised that sst2A-R expression on GHRH neurones is modulated by gonadal steroids and constitutes a mechanism for sexually differentiated GH secretion. The distribution of sst2A-R on GHRH neurones was evaluated by immunohistochemistry in adult GHRH-eGFP mice gonadectomised and treated for 3 weeks with oestradiol or testosterone implants. In gonadectomised females supplemented with testosterone, sst2A-R distribution on GHRH neurones was reduced to the level seen in intact males, whereas oestradiol implants were ineffective. Conversely, orchidectomy induced a female 'sst2A phenotype', which was reversed by testosterone supplementation. Changes in the hepatic expression of GH-dependent genes for major urinary protein-3 and the prolactin receptor reflected the altered steroid influence on GH pulsatile secretion. In the ventromedial-arcuate region, GHRH and sst2-R, as well as GHRH and somatostatin expression as measured by the real-time polymerase chain reaction, were positively correlated in both sexes. By contrast, the positive correlation between ventromedial-arcuate GHRH and periventricular somatostatin expression in males was reversed to a negative one in females. Moreover, the positive correlation between periventricular somatostatin and ventromedial-arcuate sst2-R expressions in males was lost in females. These results suggest that, in the adult mouse, testosterone is a major modulator of sst2A distribution on GHRH neurones. This marked sex difference in sst2A-R distribution may constitute a key element in the genesis of the sexually differentiated pattern of GH secretion, possibly through testosterone-modulated changes in somatostatin inputs from hypophysiotrophic periventricular neurones.


Assuntos
Estradiol/metabolismo , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Neurônios/metabolismo , Receptores de Somatostatina/metabolismo , Caracteres Sexuais , Testosterona/metabolismo , Animais , Feminino , Hormônio Liberador de Hormônio do Crescimento/genética , Hipotálamo/citologia , Hipotálamo/metabolismo , Fígado/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Orquiectomia , Ovariectomia , Receptores de Somatostatina/genética
3.
J Neuroendocrinol ; 17(3): 186-94, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15796771

RESUMO

Signal transducers and activators of transcription (STATs) are a family of transcription factors linked to class I cytokine receptors. In the present study, we investigated whether their distribution in the hypothalamus reflects the feedback regulation by growth hormone and what role they might play in the functioning of target neurones. We demonstrate that each of the seven known STATs has a distinct distribution in the hypothalamus. Notably, the STAT5 proteins, that are important in growth hormone (GH) and prolactin signalling in peripheral tissues, were expressed in somatostatin neurones of the periventricular nucleus and dopamine neurones of the arcuate nucleus. Because somatostatin neurones are regulated by feedback from circulating GH, we investigated the importance of STAT5 in these neurones. We demonstrate that STAT5b protein expression, similar to somatostatin mRNA, is sexually dimorphic in the periventricular nucleus of rats and mice. Furthermore, chronic infusion of male dwarf rats with GH increased the expression of STAT5b, while a single injection of GH into similar rats induced the phosphorylation of STAT5 proteins. The cellular abundance of somatostatin mRNA in STAT5b-deficient mice was significantly reduced in the periventricular nucleus, effectively reducing the sexually dimorphic expression. These results are consistent with the hypothesis that STAT5 proteins are involved in the feedback regulation of somatostatin neurones by GH, and that these neurones may respond to patterned GH secretion to reinforce sexual dimorphism in the GH axis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Hormônio do Crescimento/fisiologia , Hipotálamo/metabolismo , Neurônios/metabolismo , Somatostatina/metabolismo , Transativadores/fisiologia , Animais , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Nanismo Hipofisário/metabolismo , Retroalimentação Fisiológica/fisiologia , Feminino , Hormônio do Crescimento/deficiência , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Knockout , Núcleos da Linha Média do Tálamo/citologia , Núcleos da Linha Média do Tálamo/metabolismo , Proteínas do Leite/genética , Ratos , Ratos Mutantes , Fator de Transcrição STAT5 , Caracteres Sexuais , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Transativadores/deficiência , Transativadores/genética
4.
J Neuroendocrinol ; 16(1): 10-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14962070

RESUMO

Galanin-like peptide (GALP) mRNA is expressed in neurones of the hypothalamic arcuate nucleus and within pituicytes in the neurohypophysis. Several neuropeptides that are expressed in the arcuate nucleus participate in the neuroendocrine regulation of pituitary hormone secretion. Our objective was to determine the extent to which GALP might be a target for regulation by pituitary hormones or their downstream targets in the rat. The expression of GALP mRNA in the arcuate nucleus was reduced by hypophysectomy as determined by in situ hybridization. However, this did not appear to be attributable to the loss of either gonadal or adrenal steroids because castrated, ovariectomized and adrenalectomized rats had GALP mRNA expression that was indistinguishable from their respective controls. Next, we investigated the effects of growth hormone deficiency on GALP mRNA expression by studying dwarf rats and found that GALP gene expression was not different between dwarf rats and controls. We found that thyroidectomy led to a significant reduction in GALP mRNA expression compared to intact controls, and thyroidectomized rats implanted with thyroxine pellets had GALP mRNA expression that was similar to intact controls. Thus, the reduction of GALP mRNA expression seen in hypophysectomized animals may reflect, in part, a selective loss of thyroid hormone. We also found that the expression of GALP mRNA was increased in the neurohypophysis of lactating rats compared to nonlactating rats, whereas GALP mRNA expression in the arcuate nucleus was unaffected by lactation. This suggests that the induction of GALP gene expression in pituicytes is physiologically associated with activation of oxytocin and vasopressin secretion during lactation.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Peptídeo Semelhante a Galanina/metabolismo , Lactação/metabolismo , Neuro-Hipófise/metabolismo , Adrenalectomia , Animais , Castração , Peptídeo Semelhante a Galanina/genética , Hormônio do Crescimento/deficiência , Hormônio do Crescimento/fisiologia , Hipofisectomia , Lactação/genética , Hormônio Luteinizante/fisiologia , Masculino , Ovariectomia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Tireoidectomia , Tiroxina/fisiologia
5.
Endocrinology ; 144(6): 2728-40, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12746337

RESUMO

The hypothalamic GHRH neurons secrete pulses of GHRH to generate episodic GH secretion, but little is known about the mechanisms involved. We have made transgenic mice expressing enhanced green fluorescent protein (eGFP) specifically targeted to the secretory vesicles in GHRH neurons. GHRH cells transported eGFP from cell bodies in the arcuate nucleus to extensively arborized varicose fiber terminals in the median eminence. Patch clamp recordings from visually identified GHRH cells in mature animals showed spontaneous action potentials, often firing in short bursts up to 10 Hz. GHRH neurons received frequent synaptic inputs, as demonstrated by the recording of abundant inward postsynaptic currents, but spikes were followed by large after-hyperpolarizations, which limited their firing rate. Because many GHRH neurons lie close to the ventral hypothalamic surface, this was examined by wide-field binocular epifluorescence stereomicroscopy. This approach revealed an extensive horizontal network of GHRH cells at low power and individual fiber projections at higher power in the intact brain. It also showed the dense terminal projections of the GHRH cell population in the intact median eminence. This model will enable us to characterize the properties of individual GHRH neurons and their structural and functional connections with other neurons and to study directly the role of the GHRH neuronal network in generating episodic secretion of GH.


Assuntos
Núcleo Arqueado do Hipotálamo/citologia , Hormônio Liberador de Hormônio do Crescimento/genética , Neurônios/fisiologia , Potenciais de Ação/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Mapeamento Encefálico/métodos , Proteínas de Fluorescência Verde , Hibridização In Situ , Indicadores e Reagentes/metabolismo , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia de Fluorescência , Vias Neurais , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/fisiologia , RNA Mensageiro/análise
6.
J Endocrinol ; 175(2): 435-46, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12429041

RESUMO

The dwarf (dw/dw) rat differs from all other rodent models of GH deficiency in that its pituitary prolactin (PRL) content is normal or even increased. We have now studied this throughout postnatal development, using a combination of immunocytochemistry, RIA and fluorescence-activated cell sorting (FACS) and analysis. Compared with normal Albino Swiss (AS) rats, adult dw/dw rats showed a profound reduction in pituitary GH content accompanied by increased PRL content, significantly so in females (AS vs dw/dw; P<0.01). Somatotroph hypoplasia was evident in the adult dw/dw rats, with most GH(+ve) cells showing weak immunostaining, whereas many more strongly stained PRL cells were evident in pituitary sections from dw/dw rats. Facs analysis confirmed both somatotroph hypoplasia and relative lactotroph hyperplasia in dw/dw rats at all ages studied (9-144 days); the difference in somatotrophs increased with age whereas the difference in lactotrophs declined with age. At 9 days, the percentage of lactotrophs was 10-fold higher in dw/dw rats than in AS rats. Young dw/dw rats also had a higher proportion of mammosomatotrophs than AS rats, although this difference disappeared as the mammosomatotroph proportions increased with age in both strains. GHRH released GH from both dw/dw and as cells maintained in culture for 5 days. The sensitivity to GHRH and the amount of GH released was lower in the dw/dw cultures, mostly explained by their fewer GH cells and lower initial GH content. GHRH increased cAMP in as but not in dw/dw cultures, even when these were greatly enriched for dw/dw somatotrophs by FACS sorting prior to culture. These results suggest that GHRH-induced cAMP stimulation is required for trophic effects on GH synthesis and somatotroph proliferation, but is not required for GHRH-stimulated GH release. The inverse changes in somatotroph and lactotroph numbers suggest that the dw/dw mutation disturbs the mechanism that specifies and retains appropriate numbers of somatotrophs in their differentiated state, and results in a higher proportion of the remaining cells progressing to lactotrophs. The dw/dw phenotype is thus not confined to somatotrophs.


Assuntos
Diferenciação Celular/fisiologia , Adeno-Hipófise/crescimento & desenvolvimento , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , AMP Cíclico/análise , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo/métodos , Hormônio do Crescimento/análise , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Masculino , Adeno-Hipófise/citologia , Adeno-Hipófise/efeitos dos fármacos , Prolactina/análise , Radioimunoensaio , Ratos
7.
J Endocrinol ; 169(3): 519-26, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11375122

RESUMO

Cranial irradiation in children and adults often results in irreversible hypopituitarism. The earliest and most common endocrine abnormality is GH deficiency, often followed by other pituitary hormone deficits. We investigated whether a similar pattern of progressive hypopituitarism could be reproduced in an animal model. Different doses of cranial irradiation were delivered to the hypothalamo-pituitary region of normal adult male rats, and the effects on their subsequent growth, pituitary weight and hormone contents were studied. Animals received cranial irradiation with 300 kV X-rays at doses of 0, 20, 22 or 24 Gy (n=15 per group) and five animals from each group were killed at 8, 14 or 20 weeks after irradiation. Their anterior pituitary glands were weighed and assayed for GH, LH, TSH, ACTH and prolactin (PRL) content. All three doses of irradiation reduced body weight compared with that in non-irradiated controls and compromised growth between 8 and 20 weeks. Pituitary weight increased between 8 and 20 weeks in control rats, whereas it decreased significantly in the irradiated animals. Irradiation induced time- and dose-dependent changes in pituitary hormone contents. GH and PRL were most sensitive and decreased by more than 90% after irradiation; TSH contents were unaffected 8 weeks after the lowest dose of irradiation, but were reduced at 14 and 20 weeks. LH and ACTH were the slowest to be affected, and only at the greater doses of radiation. Thus progressive multiple pituitary endocrine deficits can be induced differentially in rats by increasing doses of cranial irradiation. This model should prove useful for defining the sites and mechanisms by which cranial irradiation induces neuroendocrine dysfunction.


Assuntos
Hipopituitarismo/etiologia , Sistema Hipotálamo-Hipofisário/efeitos da radiação , Lesões por Radiação/etiologia , Tolerância a Radiação , Animais , Peso Corporal/efeitos da radiação , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta à Radiação , Sistema Hipotálamo-Hipofisário/fisiopatologia , Masculino , Tamanho do Órgão/efeitos da radiação , Adeno-Hipófise/metabolismo , Adeno-Hipófise/patologia , Adeno-Hipófise/efeitos da radiação , Hormônios Hipofisários/metabolismo , Ratos , Ratos Endogâmicos
8.
Mol Cell Endocrinol ; 172(1-2): 37-45, 2001 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-11165038

RESUMO

Hypothalamic sensitivity to leptin has been suggested to be important for regulation of body fat mass. Mice heterozygous for a mutation in the leptin receptor (leptin-R) have an increased body fat mass suggesting that the abundance of leptin-R may be an important determinator of leptin sensitivity. Leptin-R cDNAs from several species contain alternative 5'untranslated regions (5'UTRs), suggesting that several distinct regulatory regions may exist. To investigate possible mechanisms by which leptin-R expression may be regulated, we searched for possible alternative 5'UTRs of the leptin-R in the rat and determined their location in relation to putative response elements. Four leptin-R 5'UTRs (exons 1A-1D), which diverged 23 bp upstream of the start codon, were identified by 5'Rapid Amplification of cDNA Ends (5'RACE) and sequencing. Exons 1B and 1C were present in 31 and 61%, respectively, of all leptin-R transcripts in the hypothalamus as determined by a ribonuclease protection assay. Analysis of the 5' flanking genomic sequences revealed an imperfect estrogen response element (ERE), two Spl-sites, three CCAAT-boxes and one octamer. Exons 1A and 1D corresponded to a putative second gene, encoding the OB-Receptor Gene Related Protein (OB-RGRP), which is transcribed from a promoter shared with the leptin-R. DNA sequencing revealed that the rat OB-RGRP had 98 and 97% homology with the mouse and human sequence, respectively. We report here that transcription of the rat leptin-R gene may generate transcripts with four alternative 5'UTRs. The presence of a putative ERE, close to the most frequently used transcriptional start sites of the leptin-R gene in the hypothalamus, provides a possible mechanism by which estrogen may exert its effects on food intake.


Assuntos
Regiões 5' não Traduzidas/análise , Proteínas de Transporte/genética , Ratos/genética , Receptores de Superfície Celular , Regiões 5' não Traduzidas/metabolismo , Animais , Sequência de Bases , Éxons , Feminino , Genoma , Peptídeos e Proteínas de Sinalização Intracelular , Dados de Sequência Molecular , Ratos Sprague-Dawley , Receptores para Leptina , Elementos de Resposta/genética , Alinhamento de Sequência , Distribuição Tecidual
9.
Hum Mol Genet ; 10(1): 39-45, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11136712

RESUMO

We have previously shown that familial septo-optic dysplasia (SOD), a syndromic form of congenital hypopituitarism involving optic nerve hypoplasia and agenesis of midline brain structures, is associated with homozygosity for an inactivating mutation in the homeobox gene HESX1/Hesx1 in man and mouse. However, as most SOD/congenital hypopituitarism occurs sporadically, the possible contribution of HESX1 mutations to the aetiology of these cases is presently unclear. Interestingly, a small proportion of mice heterozygous for the Hesx1 null allele show a milder SOD phenocopy, implying that heterozygous mutations in human HESX1 could underlie some cases of congenital pituitary hypoplasia with or without midline defects. Accordingly, we have now scanned for HESX1 mutations in 228 patients with a broad spectrum of congenital pituitary defects, ranging in severity from isolated growth hormone deficiency to SOD with panhypopituitarism. Three different heterozygous missense mutations were detected in individuals with relatively mild pituitary hypoplasia or SOD, which display incomplete penetrance and variable phenotype amongst heterozygous family members. Gel shift analysis of the HESX1-S170L mutant protein, which is encoded by the C509T mutated allele, indicated that a significant reduction in relative DNA binding activity results from this mutation. Segregation analysis of a haplotype spanning 6.1 cM, which contains the HESX1 locus, indicated that only one HESX1 mutation was present in the families containing the C509T and A541G mutations. These results demonstrate that some sporadic cases of the more common mild forms of pituitary hypoplasia have a genetic basis, resulting from heterozygous mutation of the HESX1 gene.


Assuntos
Proteínas de Homeodomínio/genética , Mutação , Nervo Óptico/anormalidades , Hipófise/anormalidades , Alelos , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Criança , Cromossomos Artificiais de Levedura , DNA/metabolismo , Saúde da Família , Feminino , Haplótipos , Heterozigoto , Humanos , Lactente , Masculino , Mutação de Sentido Incorreto , Linhagem , Fenótipo , Proteínas Repressoras , Fatores de Transcrição HES-1
10.
Endocrinology ; 141(12): 4681-9, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11108283

RESUMO

In stable transfection experiments in the GH-producing GC cell line, a construct containing the entire signal peptide and the first 22 residues of human GH linked in frame with enhanced green fluorescent protein (eGFP), produced brightly fluorescent cells with a granular distribution of eGFP. This eGFP reporter was then inserted into a 40-kb cosmid transgene containing the locus control region for the hGH gene and used to generate transgenic mice. Anterior pituitaries from these GH-eGFP transgenic mice showed numerous clusters of strongly fluorescent cells, which were also immunopositive for GH, and which could be isolated and enriched by fluorescence-activated cell sorting. Confocal scanning microscopy of pituitary GH cells from GH-eGFP transgenic mice showed a markedly granular appearance of fluorescence. Immunogold electron microscopy and RIA confirmed that the eGFP product was packaged in the dense cored secretory vesicles of somatotrophs and was secreted in parallel with GH in response to stimulation by GRF. Using eGFP fluorescence, it was possible to identify clusters of GH cells in acute pituitary slices and to observe spontaneous transient rises in their intracellular Ca2+ concentrations after loading with Ca2+ sensitive dyes. This transgenic approach opens the way to direct visualization of spontaneous and secretagogue-induced secretory mechanisms in identified GH cells.


Assuntos
Hormônio do Crescimento Humano/biossíntese , Proteínas Luminescentes/genética , Adeno-Hipófise/metabolismo , Animais , Cálcio/análise , Cosmídeos , Grânulos Citoplasmáticos/química , Citosol/química , Citometria de Fluxo , Expressão Gênica , Proteínas de Fluorescência Verde , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Hormônio do Crescimento Humano/genética , Humanos , Imuno-Histoquímica , Proteínas Luminescentes/análise , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Adeno-Hipófise/ultraestrutura
11.
Horm Res ; 53 Suppl 1: 26-33, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10895039

RESUMO

Septo-optic dysplasia (SOD) is a highly variable condition characterized by midline neurological abnormalities associated with pituitary hypoplasia and optic nerve hypoplasia. The aetiology is unknown. Mutant mice, in which a novel homeobox gene, Hesx1, has been disrupted, exhibit a phenotype that resembles the phenotype of SOD. We therefore wished to explore the possibility that this gene is implicated in SOD. We cloned and sequenced the human homologue HESX1 and screened for mutations in affected individuals using single-stranded conformational polymorphism analysis, followed by cloning and sequencing of any exons which showed a band shift. Two siblings with SOD were homozygous for an Arg53Cys missense mutation within the HESX1 homeodomain, leading to a loss of in vitro DNA binding. Subsequently, we have identified heterozygous mutations in HESX1 that are associated with milder pituitary phenotypes. Our studies indicate a vital role for Hesx1/HESX1 in forebrain and pituitary development in mouse and man, and hence in some cases of SOD.


Assuntos
Genes Homeobox , Proteínas de Homeodomínio/genética , Mutação , Nervo Óptico/anormalidades , Septo Pelúcido/anormalidades , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Humanos , Camundongos , Biologia Molecular , Linhagem , Adeno-Hipófise/embriologia , Proteínas Repressoras , Fatores de Transcrição HES-1
12.
Clin Genet ; 57(5): 337-46, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10852367

RESUMO

The development of the anterior pituitary gland is dependent upon a cascade of signalling molecules and developmental genes that function as transcription factors. Many of these genes are homeobox genes which contain a DNA-binding region or homeobox. Animal models have given a valuable insight into human pituitary disease. For example, Pit-1 and Prop1 mutants are known to have deficiencies of growth hormone, prolactin and thyroid-stimulating hormone. Human phenotypes arising as a result of mutations in these genes are similar to the mouse mutants. Mutations in the novel homeobox gene Hesx1/HESX1 are associated with the highly variable phenotype of septo-optic dysplasia in mouse and man. The unravelling of this complex developmental cascade is just commencing.


Assuntos
Deficiências do Desenvolvimento/genética , Adeno-Hipófise/crescimento & desenvolvimento , Animais , Criança , Pré-Escolar , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes Homeobox/genética , Humanos
13.
Novartis Found Symp ; 227: 206-20; discussion 220-4, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10752072

RESUMO

Despite its long-term role in postnatal growth and metabolism, pituitary growth hormone (GH) is secreted in a short-term highly episodic pulsatile pattern in all species in which it has been examined. This pattern is tightly controlled by the interplay of GH-releasing hormone (GHRH) and somatostatin (SRIF), the primary hypothalamic factors that determine GH secretion from the somatotroph and which also regulate GH synthesis and secretory reserve. The discovery of a endogenous receptor for synthetic GH secretagogues (GHS)s that differ from GHRH implies the existence of at least one other endogenous GHS system, though the physiological role of the hypothetical GHS ligand remains unclear. The GH secretory pattern is sexually dimorphic with sex differences at many levels in the hypothalamo-pituitary somatotroph axis. Studies in transgenic animals have shown that GH output is also highly sensitive to feedback control by GH itself, as well as by insulin-like growth factor I. Peripheral responses to GH are markedly dependent on the pattern of GH exposure, which is further modified after secretion by interaction with GH binding proteins and with GH receptors, both also regulated by the pattern of GH exposure. Although the hypothalamic GH pulse generator is of central importance in the control of GH output, its origin, location and mechanisms remain to be elucidated.


Assuntos
Hormônio do Crescimento/metabolismo , Receptores Acoplados a Proteínas G , Animais , Hormônio do Crescimento/fisiologia , Humanos , Receptores de Superfície Celular/metabolismo , Receptores de Grelina
14.
Growth Horm IGF Res ; 10(1): 45-52, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10753592

RESUMO

Synthetic GH-releasing peptides such as GHRP-6 are potent GH secretagogues (GHSs) in several species, but attempts to stimulate growth by continuous GHS exposure have had limited success. GHSs also release ACTH and adrenal steroids. Since glucocorticoid excess is associated with poor linear growth, stimulation of the hypothalamo-pituitary-adrenal (HPA) axis by continuous GHS administration may compromise their growth-promoting effects. We have now examined the effects of continuous GHRP-6 infusion (100 mg/day, s.c. for 14 days) in normal 150-day-old female rats, and in adrenalectomized (Adx) rats with or without dexamethasone (Dex) replacement. Infusion of GHRP-6 did not significantly affect body weight gain compared with excipient-treated controls in either intact rats (controls, 9.0 +/- 1.6 vs GHRP-6, 11.8 +/- 0.9 g) or Adx rats (4.4 +/- 1.5 vs 7.9 +/- 2.7 g). However, GHRP-6 significantly increased weight gain in Adx rats treated with Dex (controls, 3.5 +/- 1.4 vs GHRP-6, 15.4 +/- 1.6 g;P<0.01). Adrenalectomy decreased plasma triglycerides (P<0.01), and Dex treatment increased plasma cholesterol (P<0.001), GHRP-6 treatment did not affect these plasma lipids. Dex treatment also reduced plasma GH-binding protein levels and hepatic GH binding (P<0.05). Pituitary GH content was decreased in Adx rats (P<0.05) but not in Dex-treated Adx rats. Adrenalectomy markedly decreased GHS-receptor mRNA expression in the arcuate (P<0. 001) and ventromedial nuclei (P<0.01), whilst Dex treatment normalized GHS-receptor expression. These results suggest that adrenal steroids are necessary for normal GHS-receptor expression and GHRP-6-induced weight gain, but long-term stimulation of the HPA axis by continuous GHS exposure may be detrimental to the growth response.


Assuntos
Peso Corporal/efeitos dos fármacos , Glucocorticoides/fisiologia , Hormônio do Crescimento/metabolismo , Receptores da Somatotropina/metabolismo , Glândulas Suprarrenais/fisiologia , Adrenalectomia , Animais , Glicemia/metabolismo , Colesterol/sangue , Dexametasona/farmacologia , Feminino , Hormônio do Crescimento/sangue , Hipotálamo/efeitos dos fármacos , Hibridização In Situ , Insulina/sangue , Fígado/efeitos dos fármacos , Ratos , Receptores da Somatotropina/sangue , Fatores de Tempo , Triglicerídeos/sangue
15.
Clin Endocrinol (Oxf) ; 51(5): 575-85, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10594518

RESUMO

OBJECTIVES: Pulsatile GH release in humans is thought to involve the coordinated interaction of growth hormone-releasing hormone (GHRH) and somatostatin (SS). Disordered GH secretion is seen in most patients following high dose (> 30 Gy) cranial irradiation in childhood and could result from dysregulation of these hypothalamic hormones or reflect direct pituitary damage. We have used a peptide 'clamp' to assess the relative roles of continuous GHRH and intermittent SS in GH pulse generation in healthy volunteers and short-and long-term survivors of childhood brain tumours. DESIGN: Randomized controlled study. PATIENTS: 12 adult male long-term survivors of childhood brain tumours (median age 17.0 years (15.2-19. 7); 12.2 years (5.8-14.0) postradiotherapy, > 30Gy whole brain irradiation) with 9 matched control volunteers and 6 short-term survivors of childhood brain tumours (median age 6.4 years (5.9-7. 7); 2.5 years (1.7-3.6) post radiotherapy, > 30Gy whole brain irradiation) with 6 matched controls (studies of spontaneous GH release alone). MEASUREMENTS: Serum GH concentrations in 24 h spontaneous GH profiles and during three 'clamp' studies: continuous GHRH(1-29)NH2 (60 ng/kg/minutes, subcutaneous infusion, 24 h); intermittent SS(1-14) withdrawal (20microg/m2/hour, intravenous infusion, 3 h on/1 h off, 2-3 cycles over 8-12 h); intermittent SS and continuous GHRH combined (2-3 cycles over 8-12 h). Data were analysed by spectral analysis, 'peak' and 'trough' determination and serial array averaging. RESULTS: In normal adults, discrete pulsatility was seen in all profiles of spontaneous GH secretion. Continuous GHRH amplified peak GH concentrations (median basal peak 21.1 mU/l vs. GHRH 62.0 mU/l, P = 0.008) whilst pulse timing remained unaffected. Rebound GH release following SS withdrawal alone was variable. Combining continuous GHRH with intermittent SS produced regular GH responses upon SS withdrawal (20.3 mU/l; range 2. 3-105.4). Heterogeneous patterns of spontaneous GH release were seen in the irradiated subjects. Spontaneous peak GH release was reduced in the children following irradiation (Irradiation 14.9 mU/l vs. Control 25.1 mU/l, P = 0.007). Peak GH concentrations were significantly amplified by GHRH in half of them. Adult long-term survivors had lower spontaneous GH concentrations and continuous GHRH amplified GH release in most subjects (Spontaneous 4.2 mU/l vs. GHRH 6.5 mU/l, P = 0.008) but peak concentrations remained far less than those of controls. Combining intermittent SS with continuous GHRH regularized GH release in many patients but the GH responses remained attenuated (4.6 mU/l; 2.5-17.5). CONCLUSION: GH pulsatility can be generated in normal volunteers by the combination of continuous GHRH and intermittent SS and provides indirect evidence for a role for GHRH in GH synthesis and replenishment of stored GH pools at times of high SS tone. Patterns of GH release in short-and long-term survivors of childhood brain tumours are heterogeneous suggesting that combined hypothalamic deficiencies of GHRH and SS occur following high dose radiotherapy. The attenuated GH release seen in long-term survivors compared to controls suggests that GH secretory dysfunction does not simply reflect reduced GHRH and SS secretion, and that trophic effects or pituitary damage may be important with time.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/radioterapia , Irradiação Craniana , Sermorelina , Somatostatina/metabolismo , Adolescente , Adulto , Neoplasias Encefálicas/sangue , Estudos de Casos e Controles , Humanos , Masculino , Taxa Secretória , Somatostatina/sangue , Estatísticas não Paramétricas , Fatores de Tempo
16.
Clin Endocrinol (Oxf) ; 50(3): 301-8, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10435054

RESUMO

OBJECTIVE: Growth hormone (GH) is secreted in a pulsatile fashion promoting growth and a number of diverse metabolic actions. The precise components of the pulsatile signal involved in growth regulation are unclear. DESIGN: A retrospective analysis of 24 h serum GH concentration profiles to evaluate the relative contribution of peak and trough serum GH concentrations to growth regulation, GH response to insulin induced hypoglycaemia (ITT) and serum insulin like growth factor-1 (IGF-1) concentration. PATIENTS: Fifty short prepubertal children (age 5.2-11.9 years). MEASUREMENT: Analysis of the hormone profile by a concentration distribution method that determines the concentration at or below which the serum GH concentrations in the 24 hour profile spend a percentage of the total time. The method generates an estimate of the observed concentrations (OC) below which 95% and 5% of the values in the time series lie: OC 95 (peaks) and OC5 (troughs). RESULTS: Twenty six of the children were growing at a normal rate for short children with a height velocity standard deviation score (HVSDS) between +0.4 and -0.8 whereas twenty four were growing more slowly (HVSDS between -0.9 to -3.9). The former group had a mean peak GH response to ITT of 27.3 (11.1) mU/l whereas the latter had a mean value of 8.7 (6.5) mU/l. There was no relationship between (peak and trough GH concentration) and the age of the individual or body mass index. Peak GH levels were positively related to HVSDS and serum IGF-1 values (r = 0.44; P = 0.002 and r = 0.53; P = 0.002, respectively). GH trough levels were inversely related to these measurements (r = -0.29; P = 0.05; and r = -0.46; P = 0.002, respectively). Further analysis showed that individuals with the slowest growth rates and lowest IGF-1 concentrations had the lowest peak and highest trough GH concentrations (ANOVA F = 6.0; P = 0.002). Similarly, the peak GH response to ITT was lowest in those individuals with high troughs and low peaks (ANOVA F = 9.99; P < 0.001). CONCLUSIONS: These results suggest that the peak values of a GH concentration profile influence growth rate and the IGF-1 axis whereas elevated trough values have the greatest influence on growth rate and IGF-1 values when GH peaks are low.


Assuntos
Transtornos do Crescimento/sangue , Hormônio do Crescimento/sangue , Crescimento/fisiologia , Fator de Crescimento Insulin-Like I/análise , Análise de Variância , Criança , Pré-Escolar , Feminino , Humanos , Insulina/sangue , Masculino , Estudos Retrospectivos
17.
Neuroendocrinology ; 69(6): 417-23, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10364693

RESUMO

We have demonstrated previously that pharmacological doses of oestradiol decreased leptin receptor expression in the hypothalamus. We therefore analysed leptin receptor expression during the oestrous cycle in the rat, to establish if acute changes in oestradiol affect leptin receptor expression under physiological conditions. Radioactive in situ hybridization histochemistry was used to measure the gene expression under investigation. Total leptin receptor transcript levels were lowest in pro-oestrus in the choroid plexus, these changes correspond inversely with levels of circulating oestradiol in the rat 4-day oestrous cycle. In contrast full-length leptin receptor levels in both arcuate and ventromedial nuclei did not correspond to the levels of total leptin receptor in the same areas of the hypothalamus or serum levels of oestradiol. Full-length leptin receptor expression in the arcuate nucleus was negatively correlated with neuropeptide Y (NPY) expression (r = 0.447, p < 0. 05) in the same nucleus. Arcuate nucleus NPY expression did not correlate significantly with the expression of total leptin receptors in the arcuate nucleus (r = 0.080) or serum leptin levels (r = 0.251). Our results demonstrate that leptin receptor expression is regulated during the oestrous cycle. The unchanged serum leptin levels during the oestrous cycle together with the correlation between the expression of leptin-RL and NPY provide circumstantial evidence that regulation of leptin receptor abundance in the hypothalamus governs the biological actions of leptin.


Assuntos
Proteínas de Transporte/metabolismo , Estro/metabolismo , Leptina/metabolismo , Neuropeptídeo Y/biossíntese , Receptores de Superfície Celular , Animais , Peso Corporal/fisiologia , Química Encefálica/fisiologia , DNA Complementar/biossíntese , Estradiol/biossíntese , Feminino , Hibridização In Situ , Progesterona/biossíntese , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos , Receptores para Leptina
18.
J Neuroendocrinol ; 11(3): 229-36, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10201819

RESUMO

In transgenic growth-retarded (Tgr) rats, expression of human growth hormone (hGH) is targeted to hypothalamic GH-releasing hormone (GHRH) neurones. In these rats, GHRH is reduced and somatostatin expression is increased, resulting in secondary GH deficiency and dwarfism. Tgr rats also show reduced pituitary prolactin (PRL), which may reflect an additional lactogenic feedback action of the hGH transgene, analogous to that in mice transgenic for peripheral hGH which show enhanced dopamine (DA) and tyrosine hydroxylase (TH) expression in the hypothalamic tuberoinfundibular dopaminergic (TIDA) neurones that inhibit PRL secretion. The present study examined DA histofluorescence and TH immunoreactivity in Tgr rats, and also in dw/dw rats, a dwarf strain with primary pituitary GH but not PRL deficiency. Radioimmunoassay confirmed a significant decrease in total pituitary PRL content in Tgr rats, but showed a marked increase in total pituitary PRL in dw/dw rats. Despite their PRL deficiency, Tgr rats showed qualitatively increased TIDA histofluorescence and TH immunoreactivity compared with AS control rats, though the total number of detectable TH-positive TIDA neurones was similar for Tgr and AS. In contrast, dw/dw rats showed increased numbers of TH-immunoreactive TIDA neurones whilst TIDA fluorescence was unchanged, and these findings were not affected in dw/dw rats given bovine GH (200 microg/d s.c. for 7 d). These results suggest that reduced PRL in Tgr rats is due to a local lactogenic feedback effect of hGH to stimulate TIDA neurones. The complex changes in TIDA neurones probably reflect a combination of increased lactogenic feedback in Tgr rats, with an increased (Tgr) or decreased (dw/dw) somatogenic feedback on GHRH neurones, some of which coexpress TH. Thus, the unchanged number of TIDA neurones in Tgr rats may result from hGH stimulation of TH and DA, but a reduction in GHRH-producing cells, whereas increased TIDA neurones in dw/dw rats suggests a stimulation by endogenous PRL with an increased GHRH cell complement due to GH deficiency. These findings therefore indicate that differences in lactogenic feedback in these dwarf rat models are reflected in marked differences in their hypothalamic TIDA neurones.


Assuntos
Dopamina/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Hipófise/metabolismo , Prolactina/metabolismo , Animais , Animais Geneticamente Modificados , Hormônio do Crescimento/genética , Hipotálamo/citologia , Masculino , Modelos Biológicos , Radioimunoensaio , Ratos , Ratos Mutantes
19.
Acta Paediatr Suppl ; 88(433): 49-54, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10626545

RESUMO

The homeobox gene Hesx1, which encodes a pituitary transcription factor, is first expressed at gastrulation in the mouse embryo. Hesx1 expression begins in prospective forebrain tissue but later becomes restricted to Rathke's pouch, the primordium of the anterior pituitary gland. Transgenic mice lacking Hesx1 exhibit a phenotype comprising variable anterior CNS defects, such as a reduced prosencephalon, abnormalities in the corpus callosum and septum pellucidum, anophthalmia or microphthalmia, defective olfactory development and bifurcations in Rathke's pouch with pituitary dysplasia. A comparable and highly variable phenotype in humans is septo-optic dysplasia. We have cloned and sequenced the human homologue HESX1 and screened for mutations in affected individuals using single-stranded conformational polymorphism analysis. Two siblings with septo-optic dysplasia were homozygous for a missense mutation within the HESX1 homeobox. This mutation resulted in the substitution of a highly conserved arginine residue (Arg53) by cysteine and led to a loss of in vitro DNA binding. Hence, a vital role for Hesx1/HESX1 in forebrain and pituitary development in mice and humans is suggested.


Assuntos
Genes Homeobox , Sequências Hélice-Alça-Hélice/genética , Proteínas de Homeodomínio/genética , Septo Pelúcido/anormalidades , Animais , Arginina/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Cisteína/genética , Genótipo , Humanos , Mutação de Sentido Incorreto , Fenótipo , Adeno-Hipófise/fisiologia , Prosencéfalo/fisiologia , Proteínas Repressoras , Fatores de Transcrição HES-1 , Transcrição Gênica
20.
Mol Pharmacol ; 54(5): 864-73, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9804621

RESUMO

Somatostatin (SRIF) is the main inhibitory peptide regulating growth hormone (GH) secretion. It has been difficult to establish the role of endogenous SRIF release in the absence of pure SRIF antagonists. Although several SRIF antagonists have recently been described, none have been shown to possess in vivo activity in the absence of added SRIF. Here, an SRIF antagonist with no detectable agonist activity has been identified from a synthetic combinatorial hexapeptide library containing 6.4 x 10(7) unique peptides. Each peptide in the library is amino-terminally acetylated and carboxyl-terminally amidated and consists entirely of D-amino acids. A SRIF-responsive yeast growth assay was used as a primary screening tool, and cAMP accumulation, competitive binding, and microphysiometry also were used to confirm and further characterize SRIF antagonist activity. The hexapeptide library was screened in stepwise iterative fashion to identify AC-178,335, a pure SRIF antagonist of the sequence Ac-hfirwf-NH2. This D-hexapeptide bound SRIF receptor type 2 with an affinity constant (Ki) of 172 +/- 12 nM, blocked SRIF inhibition of adenylate cyclase in vitro (IC50 = 5.1 +/- 1.4 microM), and induced GH release when given alone (50 micrograms intravenously) to anesthetized rats with or without pretreatment with a long-acting SRIF agonist.


Assuntos
Hormônio do Crescimento/metabolismo , Oligopeptídeos/farmacologia , Somatostatina/antagonistas & inibidores , Somatostatina/fisiologia , Animais , Ligação Competitiva , Linhagem Celular , AMP Cíclico/metabolismo , Masculino , Oligopeptídeos/metabolismo , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/antagonistas & inibidores , Receptores de Somatostatina/metabolismo , Saccharomyces cerevisiae/efeitos dos fármacos , Saccharomyces cerevisiae/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...