Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
J Endocrinol ; 229(3): 331-41, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27029473

RESUMO

Pituitary follicle-stimulating hormone (FSH) synthesis is regulated by transforming growth factorßsuperfamily ligands, most notably the activins and inhibins. Bone morphogenetic proteins (BMPs) also regulate FSHß subunit (Fshb) expression in immortalized murine gonadotrope-like LßT2 cells and in primary murine or ovine primary pituitary cultures. BMP2 signals preferentially via the BMP type I receptor, BMPR1A, to stimulate murine Fshb transcription in vitro Here, we used a Cre-lox approach to assess BMPR1A's role in FSH synthesis in mice in vivo Gonadotrope-specific Bmpr1a knockout animals developed normally and had reproductive organ weights comparable with those of controls. Knockouts were fertile, with normal serum gonadotropins and pituitary gonadotropin subunit mRNA expression. Cre-mediated recombination of the floxed Bmpr1a allele was efficient and specific, as indicated by PCR analysis of diverse tissues and isolated gonadotrope cells. Furthermore, BMP2 stimulation of inhibitor of DNA binding 3 expression was impaired in gonadotropes isolated from Bmpr1a knockout mice, confirming the loss of functional receptor protein in these cells. Treatment of purified gonadotropes with small-molecule inhibitors of BMPR1A (and the related receptors BMPR1B and ACVR1) suppressed Fshb mRNA expression, suggesting that an autocrine BMP-like molecule might regulate FSH synthesis. However, deletion of Bmpr1a and Acvr1 in cultured pituitary cells did not alter Fshb expression, indicating that the inhibitors had off-target effects. In sum, BMPs or related ligands acting via BMPR1A or ACVR1 are unlikely to play direct physiological roles in FSH synthesis by murine gonadotrope cells.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/deficiência , Fertilidade/fisiologia , Gonadotrofos/fisiologia , Gonadotropinas Hipofisárias/biossíntese , Receptores de Ativinas Tipo I/deficiência , Receptores de Ativinas Tipo I/genética , Receptores de Ativinas Tipo I/fisiologia , Animais , Proteína Morfogenética Óssea 2/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/antagonistas & inibidores , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Células Cultivadas , Feminino , Subunidade beta do Hormônio Folículoestimulante/biossíntese , Subunidade beta do Hormônio Folículoestimulante/genética , Gonadotrofos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
2.
Neurol India ; 62(3): 280-4, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25033850

RESUMO

OBJECTIVE: To study the dynamic changes of pituitary hormones in traumatic brain injury (TBI) and to correlate the severity and neurological outcome. PATIENTS AND METHODS: Dynamic changes in the pituitary hormones were evaluated in 164 patients with TBI on day-1, day-7, day-14, day-21, and day-28 post injury. Admission TBI severity and long-term outcome were assessed with Glasgow Coma Scale (GCS) score and Glasgow Outcome Scale (GOS) score. The pituitary hormonal changes were correlated with TBI severity and outcome. RESULTS: Of the 164 patients included in the study, pituitary dysfunction was found in 84 patients and in the remaining 80 patients pituitary function was normal. Most of the pituitary hormone deficiencies observed resolved over time; however, a significant proportion of patients had pituitary dysfunction at one month post injury. The hormones associated with poor outcome included growth hormone, thyrotropic hormone, and gonadotropic hormone. CONCLUSION: Dynamic changes of pituitary hormones in patients with TBI may reflect the severity of injury and also determine the outcome. Deficiency of growth hormone, gonadotropic hormone, and thyrotropic hormone can adversely affect neurological outcome.


Assuntos
Lesões Encefálicas/complicações , Doenças da Hipófise/etiologia , Hormônios Hipofisários/biossíntese , Adulto , Feminino , Escala de Coma de Glasgow , Escala de Resultado de Glasgow , Gonadotropinas Hipofisárias/biossíntese , Hormônio do Crescimento Humano/biossíntese , Humanos , Hipopituitarismo/etiologia , Hipopituitarismo/metabolismo , Masculino , Pessoa de Meia-Idade , Avaliação de Resultados da Assistência ao Paciente , Doenças da Hipófise/metabolismo , Índice de Gravidade de Doença , Tireotropina/biossíntese
3.
J Clin Invest ; 122(10): 3791-5, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23006326

RESUMO

Hyperprolactinemia is the most common cause of hypogonadotropic anovulation and is one of the leading causes of infertility in women aged 25-34. Hyperprolactinemia has been proposed to block ovulation through inhibition of GnRH release. Kisspeptin neurons, which express prolactin receptors, were recently identified as major regulators of GnRH neurons. To mimic the human pathology of anovulation, we continuously infused female mice with prolactin. Our studies demonstrated that hyperprolactinemia in mice induced anovulation, reduced GnRH and gonadotropin secretion, and diminished kisspeptin expression. Kisspeptin administration restored gonadotropin secretion and ovarian cyclicity, suggesting that kisspeptin neurons play a major role in hyperprolactinemic anovulation. Our studies indicate that administration of kisspeptin may serve as an alternative therapeutic approach to restore the fertility of hyperprolactinemic women who are resistant or intolerant to dopamine agonists.


Assuntos
Anovulação/tratamento farmacológico , Hiperprolactinemia/tratamento farmacológico , Kisspeptinas/uso terapêutico , Animais , Anovulação/etiologia , Anovulação/fisiopatologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Ciclo Estral/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas Hipofisárias/biossíntese , Gonadotropinas Hipofisárias/sangue , Gonadotropinas Hipofisárias/metabolismo , Hiperprolactinemia/induzido quimicamente , Hiperprolactinemia/complicações , Hiperprolactinemia/fisiopatologia , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/fisiologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Bombas de Infusão Implantáveis , Kisspeptinas/biossíntese , Kisspeptinas/genética , Masculino , Camundongos , Prolactina/administração & dosagem , Prolactina/toxicidade , Fluxo Pulsátil , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
4.
J Mol Neurosci ; 48(3): 603-16, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22399226

RESUMO

In vertebrates, gonadotropin-releasing hormone (GnRH) and pituitary adenylate cyclase-activating polypeptide (PACAP) are key hormones regulating growth and reproduction in the brain-pituitary axis. The regulating hormonal interactions are of great interest, therefore, the aim of this study is to provide novel insights into the involvement of brain GnRH and PACAP in oogensis and spermatogenesis in a fish model, the blue gourami (Trichogaster trichopterus). cDNA cloning of two GnRH forms combined with phylogenetic analysis revealed that three paralogous GnRH forms exist in blue gourami and evolve as a result of genome duplication. GnRH1 mRNA levels are related to final oocyte maturation (FOM), and this peptide stimulated ß follicle-stimulating hormone (ßFSH) and growth hormone (GH) gene expression; GnRH2 stimulated ß gonadotropins (GtH) gene expression and GnRH analog combined with PACAP-38 synergistically upregulate GH and ßFSH gene expression. The data presented, together with previous studies in our lab, enable suggesting mechanisms explaining the physiological relevance of these peptides in the regulation of gametogenesis and steroidogenesis in blue gourami females. These findings support the biological importance of the GnRH and PACAP hormones family, enabling them to stimulate differential biological functions in the regulation of growth and reproduction.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Gonadotropinas Hipofisárias/biossíntese , Oogênese/fisiologia , Fragmentos de Peptídeos/fisiologia , Perciformes/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Precursores de Proteínas/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , DNA Complementar/genética , Sinergismo Farmacológico , Feminino , Peixes/genética , Duplicação Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas Hipofisárias/genética , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Perciformes/genética , Perciformes/crescimento & desenvolvimento , Filogenia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Hipófise/citologia , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Precursores de Proteínas/genética , Precursores de Proteínas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Alinhamento de Sequência , Homologia de Sequência , Especificidade da Espécie , Vitelogênese/fisiologia
5.
Endocrinology ; 153(5): 2332-43, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22374973

RESUMO

A neuropeptide that directly inhibits gonadotropin secretion from the pituitary was discovered in quail and named gonadotropin-inhibitory hormone (GnIH). The presence and functional roles of GnIH orthologs, RF-amide-related peptides (RFRP), that possess a common C-terminal LPXRF-amide (X = L or Q) motif have also been demonstrated in mammals. GnIH orthologs inhibit gonadotropin synthesis and release by acting on pituitary gonadotropes and GnRH neurons in the hypothalamus via its receptor (GnIH receptor). It is becoming increasingly clear that GnIH is an important hypothalamic neuropeptide controlling reproduction, but the detailed signaling pathway mediating the inhibitory effect of GnIH on target cells is still unknown. In the present study, we investigated the pathway of GnIH cell signaling and its possible interaction with GnRH signaling using a mouse gonadotrope cell line, LßT2. First, we demonstrated the expression of GnIH receptor mRNA in LßT2 cells by RT-PCR. We then examined the inhibitory effects of mouse GnIH orthologs [mouse RFRP (mRFRP)] on GnRH-induced cell signaling events. We showed that mRFRP effectively inhibited GnRH-induced cAMP signaling by using a cAMP-sensitive reporter system and measuring cAMP levels, indicating that mRFRP function as an inhibitor of adenylate cyclase. We further showed that mRFRP inhibited GnRH-stimulated ERK phosphorylation, and this effect was mediated by the inhibition of the protein kinase A pathway. Finally, we demonstrated that mRFRP inhibited GnRH-stimulated gonadotropin subunit gene transcriptions and also LH release. Taken together, the results indicate that mRFRP function as GnIH to inhibit GnRH-induced gonadotropin subunit gene transcriptions by inhibiting adenylate cyclase/cAMP/protein kinase A-dependent ERK activation in LßT2 cells.


Assuntos
Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Gonadotropinas Hipofisárias/biossíntese , Neuropeptídeos/farmacologia , Transdução de Sinais/fisiologia , Adenilil Ciclases/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gonadotrofos/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas Hipofisárias/genética , Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/metabolismo , Camundongos , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/química , Neuropeptídeos/genética , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica
6.
J Endocrinol ; 191(1): 309-17, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17065413

RESUMO

Depending on the estrous/menstrual cycle stage in females, ovarian-derived estradiol (E(2)) exerts either a negative or a positive effect on the hypothalamic-pituitary axis to regulate the synthesis and secretion of pituitary gonadotropins, LH, and FSH. To study the role of estrogen receptor-alpha (ERalpha) mediating these effects, we assessed the relevant parameters in adult wild-type (WT) and ERalpha-null (alphaERKO) female mice in vivo and in primary pituitary cell cultures. The alphaERKO mice exhibited significantly higher plasma and pituitary LH levels relative to WT females despite possessing markedly high levels of circulating E(2). In contrast, hypothalamic GnRH content and circulating FSH levels were comparable between genotypes. Ovariectomy led to increased plasma LH in WT females but no further increase in alphaERKO females, while plasma FSH levels increased in both genotypes. E(2) treatment suppressed the high plasma LH and pituitary Lhb mRNA expression in ovariectomized WT females but had no effect in alphaERKO. In contrast, E(2) treatments only partially suppressed plasma FSH in ovariectomized WT females, but this too was lacking in alphaERKO females. Therefore, negative feedback on FSH is partially E(2)/ERalpha mediated but more dependent on ovarian-derived inhibin, which was increased threefold above normal in alphaERKO females. Together, these data indicate that E(2)-mediated negative feedback is dependent on functional ERalpha and acts to primarily regulate LH synthesis and secretion. Studies in primary cultures of pituitary cells from WT females revealed that E(2) did not suppress basal or GnRH-induced LH secretion but instead enhanced the latter response, indicating that the positive influence of E(2) on gonadotropin secretion may occur at the level of the pituitary. Once again this effect was lacking in alphaERKO gonadotropes in culture. These data indicate that the aspects of negative and positive effects of E(2) on gonadotropin secretion are ERalpha dependent and occur at the level of the hypothalamus and pituitary respectively.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Gonadotropinas Hipofisárias/biossíntese , Hipotálamo/metabolismo , Hipófise/metabolismo , Animais , Northern Blotting/métodos , Células Cultivadas , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas Hipofisárias/metabolismo , Hormônio Luteinizante/biossíntese , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Hormônio Luteinizante Subunidade beta/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovariectomia , Hipófise/química , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Reproduction ; 128(1): 43-52, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15232063

RESUMO

Measurement of inhibins A and B in the serum of normal cyclic rodents has implicated FSH in the regulation of these peptides within the ovary. To extend these observations we have used a panel of mutant mice carrying mutations which affect either the production of, or the ability to respond to, FSH and LH. As a consequence, the females are infertile and show different degrees of follicular development. The aim of this study was to measure inhibin gene transcription in the ovaries of these mutant females together with inhibin protein levels in ovaries and serum and to relate these to follicular development within the ovary. Comparison was made with a pool of normal/heterozygous females. In hpg females where lack of GnRH production results in the absence of gonadotropin synthesis, in FSHbeta knockout (FSHbetaKO) females where disruption of the gene encoding FSHbeta results in the absence of FSH production, and in FSH receptor knockout (FSHRKO) females which are unable to respond to circulating FSH, follicular development remains at the pre-antral stage in these three mutants. Only in the hpg females were common inhibin alpha subunit mRNA levels significantly lower than normal. In these three mutants, however, mRNA levels for both the betaA and betaB subunits were extremely low compared with normal mice. At the protein level, neither inhibin A nor B was detected in the serum of these three mutants; however inhibin B, albeit at very low levels, was detectable within the ovaries. These observations confirm a major role for FSH in the control of transcription of the betaA and betaB genes but suggest that the constitutive transcription of the alpha subunit is less dependent on FSH. In contrast, in LH receptor knockout (LuRKO) female mice inhibin betaA subunit mRNA levels were similar to those measured in normal/heterozygous females but levels of inhibin alpha and betaB subunit mRNAs were significantly higher than in the normal group. This was reflected in significantly higher inhibin B protein levels in ovaries and serum. An inability to respond to LH combined with high circulating levels of FSH leads to a high proportion of antral follicles in LuRKO females, with granulosa cells constituting the major cell type within the ovary. The high percentage of antral granulosa cells is likely to account for the significantly higher levels of inhibin B production in these ovaries.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Gonadotropinas Hipofisárias/biossíntese , Subunidades beta de Inibinas/genética , Ovário/metabolismo , RNA Mensageiro/metabolismo , Animais , Feminino , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/sangue , Subunidade beta do Hormônio Folículoestimulante/genética , Gonadotropinas Hipofisárias/metabolismo , Imuno-Histoquímica/métodos , Subunidades beta de Inibinas/análise , Subunidades beta de Inibinas/sangue , Inibinas/análise , Inibinas/sangue , Inibinas/genética , Hormônio Luteinizante/biossíntese , Hormônio Luteinizante/sangue , Camundongos , Camundongos Knockout , Camundongos Mutantes , Mutação , Folículo Ovariano/fisiologia , Ovário/química , RNA Mensageiro/análise , Transcrição Gênica
10.
J Theor Biol ; 218(4): 457-70, 2002 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-12384049

RESUMO

We analyse computational modules of a frequency decoding signal transduction network. The gonadotropin releasing hormone (GnRH) signal transduction network mediates the biosynthesis and release of the gonadotropins, luteinizing hormone (LH) and follicle stimulating hormone (FSH). The pulsatile pattern of GnRH production by the hypothalamus has a critical influence on the release and synthesis of gonadotropins in the pituitary. In humans, slower pulses lead to the expression of the beta-subunit of the LH protein and cause anovulation and amenorrhea. Higher frequency pulses lead to expression of the alpha subunit and a hypogonadal state. The frequency sensitivity is a consequence of the structure of the GnRH signal transduction network. We analyse individual components of this network, organized into three network architectures, and describe the frequency-decoding capabilities of each of these modules. We find that these modules are comparable to simple circuit elements, some of which integrate and others which perform as frequency sensitive filters. We propose that the cell computes by exploiting variation in the time scales of protein activation (phosphorylation) and gene expression.


Assuntos
Simulação por Computador , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas Hipofisárias/metabolismo , Transdução de Sinais/fisiologia , Animais , Retroalimentação , Gonadotropinas Hipofisárias/biossíntese , Hipotálamo/metabolismo , Modelos Biológicos , Fosforilação , Hipófise/metabolismo , Fatores de Tempo
11.
Gen Comp Endocrinol ; 121(2): 135-45, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11178879

RESUMO

The objective of the present study was to investigate the in vivo effects of sex steroids (estradiol-17beta, E(2); testosterone, T) and the nonaromatizable androgen 5alpha-dihydrotestosterone (DHT) on the levels of gonadotropin II (GTH II) in plasma and pituitary and on aromatase activity in 2-year-old male black porgy, Acanthopagrus schlegeli, during the prereproductive season. Black porgy GTH II and GTH II beta subunits were purified and anti-GTH II beta serum was induced. A specific radioimmunoassay for black porgy GTH II was developed. cDNA GTH II beta was also cloned from a black porgy pituitary cDNA library for use as a probe for Northern analysis. Male fish were divided into eight groups (n = 64): control; E(2) (3 doses, 2.4 ng, 72 ng, and 2.2 microg/g body weight); T (2 doses, 72 ng and 2.2 microg/g body weight); and DHT (2 doses, 72 ng and 2.2 microg/g body weight). Fish were injected with the respective vehicle or different doses of material on days 0, 8, and 16. Plasma was collected at 4-day intervals from days 4 to 20. Plasma GTH II concentrations were significantly increased (up to 45-fold) in the E(2) group from days 4 to 20 in a dose-dependent manner. In a further experiment during the late reproductive season, plasma GTH II levels increased at 4 h and on days 1 and 2 following a single injection of 1.0 microg E(2)/g body weight (on day 0). Androgens (T or DHT) had little or no effect on plasma GTH II. Pituitary GTH II contents on day 20 were significantly lower in the 72-ng E(2) and 2.2-microg E(2) groups but not in the 2.4-ng E(2) group compared with the control group. Pituitary GTH II beta mRNA levels were significantly stimulated in the 72-ng and 2.2-microg E(2) groups on day 20. Gonadal aromatase activity was not significantly changed in any of the treated or control groups. It is concluded that GTH II secretion in black porgy is regulated by an estrogen-specific effect. Increased plasma GTH II levels or other factors in addition to E(2) might be involved in the regulation of gonadal aromatase activity and sex change in protandrous black porgy.


Assuntos
Estradiol/farmacologia , Gonadotropinas Hipofisárias/biossíntese , Perciformes/metabolismo , Sequência de Aminoácidos , Animais , Aromatase/metabolismo , Sequência de Bases , Northern Blotting , Di-Hidrotestosterona/metabolismo , Relação Dose-Resposta a Droga , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Gonadotropinas Hipofisárias/metabolismo , Organismos Hermafroditas , Masculino , Dados de Sequência Molecular , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , RNA Mensageiro/biossíntese , Radioimunoensaio , Processos de Determinação Sexual , Testosterona/farmacologia
12.
J Exp Zool ; 286(1): 64-75, 2000 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-10607369

RESUMO

A study was carried out in tilapia in order to see whether the gonadotropin (GtH) beta subunits show distinct patterns of expression at different stages of their reproductive development. Male and female tilapia hybrids (Oreochromis niloticus x O. aureus) were collected at various times of the year, and a number of parameters were measured in order to establish the reproductive state of the fish. Circulating testosterone (T), estradiol (E(2)) and 11 ketotestosterone (11KT) levels were assayed, gonads were removed for calculation of gonadosomatic index (GSI) values and histological studies, and RNA was extracted from the pituitaries for measurement of GtH Ibeta and IIbeta mRNA levels. In maturing fish of both sexes, the circulating steroid levels were positively correlated with each other (r(2) = 0.66-0.91) and in males, also with the GSI values (r(2) = 0.68). A positive correlation was also seen in these fish between GSI values and the prevalence of spermatocytes and spermatids (r(2) = 0.54). In maturing females, the maximal oocyte diameter was positively correlated with circulating E(2) levels (r(2) = 0.63), while GSI values showed no correlation; this presumably relates to the cycling nature of this asynchronous spawner. In regressing fish of both sexes, no clear correlation between these reproductive parameters was seen. In all fish, the GtH Ibeta mRNA levels were highest in fish with steroids ranging 1-10 ng T or E(2)/ml for males or females, respectively, and were lower in fish with steroids at higher or lower levels. In fish with high steroid levels, the IIbeta mRNA levels were also high, and in regressed males the increases were positively correlated. Exposure of cultured pituitary cells to either steroid (T at >10 nM, or E(2) at >1 nM) was followed by a decrease in the steady-state levels of the Ibeta transcript, while those of IIbeta were left unaltered. In situ hybridization studies revealed that in pituitaries of both sexes, the cells producing each of these mRNAs are located in a distinct location. These results suggest that gonadal steroids may exert differential feedback mechanisms at the level of the pituitary to control transcription of each GtH beta subunit in distinct cell types specific for each hormone.


Assuntos
Gonadotropinas Hipofisárias/genética , RNA Mensageiro/metabolismo , Reprodução , Maturidade Sexual , Tilápia/crescimento & desenvolvimento , Animais , Estradiol/sangue , Feminino , Gonadotropinas Hipofisárias/biossíntese , Hibridização In Situ , Masculino , Testosterona/sangue , Tilápia/sangue
13.
J Neuroendocrinol ; 11(10): 785-94, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10520127

RESUMO

In addition to regulating anterior pituitary function by being released from the median eminence, mammalian neurotensin (NT) may also exert an autocrine or a paracrine action within the anterior pituitary. In this study, using double immunostaining with elution restaining, we identified the specific anterior pituitary cells which express NT immunoreactivity (NT-IR) during the rat oestrous cycle. In the normal cycling rat, NT-IR was present in both gonadotrophs and thyrotrophs and displayed plastic changes along the oestrous cycle. Both the number of TSH-NT positive cells and the intensity of immunological reaction were elevated during dioestrus, and decreased through pro-oestrus and early oestrus. NT-IR was also high in both follicle stimulating hormone (FSH)- or luteinizing hormone (LH)-positive cells during early pro-oestrus, and decreased during late pro-oestrus. Treatment of intact rats with either the anti-oestrogens Tamoxifen or LY117018, or the anti-progestagen RU486 prevented the normal expression of NT-IR in thyroid-stimulating hormone (TSH)-, FSH-, and LH-positive cells during pro-oestrus. Bilateral ovariectomy induced a dramatic reduction in the number of NT-IR cells. This effect was completely prevented by treatment of ovariectomized rats with oestradiol and progesterone, and was unaffected by the concurrent administration of a GnRH antagonist. Furthermore, administration of an anti-oestrogen together with an anti-progestagen to ovariectomized-oestrogen, progesterone-treated rats, blocked the stimulatory effect of ovarian hormones on NT-IR in anterior pituitary cells. These findings demonstrate that, in female rats, NT is specifically localized in gonadotrophs or thyrotrophs. In addition, they strongly suggest that changes in circulating concentrations of ovarian steroids may control both NT synthesis in, and release from, these cells.


Assuntos
Estradiol/fisiologia , Gonadotropinas Hipofisárias/biossíntese , Neurotensina/biossíntese , Adeno-Hipófise/metabolismo , Progesterona/fisiologia , Tireotropina/biossíntese , Animais , Moduladores de Receptor Estrogênico/farmacologia , Estro/efeitos dos fármacos , Estro/fisiologia , Feminino , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Imuno-Histoquímica , Hormônio Luteinizante/metabolismo , Ovariectomia , Adeno-Hipófise/citologia , Adeno-Hipófise/fisiologia , Progesterona/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
14.
Arch Dis Child ; 80(4): 324-9, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10086936

RESUMO

OBJECTIVE: To study the value of measuring serum luteinising hormone (LH), follicle stimulating hormone (FSH), testosterone, and dihydrotestosterone (DHT) in androgen insensitivity syndrome (AIS). DESIGN: Retrospective study of patients on a nationwide register of AIS. PATIENTS: Sixty one cases of AIS with androgen receptor (AR) dysfunction (abnormalities of the AR gene and/or abnormal AR binding) were divided into three age groups: infants, < 1 year old; children, 1-13 years old; and postpubertal, > 13 years old. MEASUREMENTS: Age, dose of human chorionic gonadotrophin (hCG) stimulation, pre-hCG and post-hCG serum testosterone values, serum DHT values, and serum LH and FSH values before and after LH releasing hormone (LHRH) stimulation. RESULTS: In 23 of 30 infants testosterone was within age related reference ranges; six were above this range. The median testosterone rise following variable dosage of hCG was 9.5 times the basal value. The increment was not related to the hCG dose, age, or basal concentration of testosterone. The median basal and stimulated testosterone:DHT ratios were 2.5 and 6.1, respectively. The median increment in DHT was 2.2-fold. Seventeen of 18 FSH and 11 of 19 LH measurements were within age related ranges in infants; in seven patients LH values were above the range. LHRH stimulation performed in 39 patients showed an exaggerated LH in all age groups. The FSH response was not exaggerated in children. CONCLUSION: Although a positive hCG test excludes biosynthetic defects of testosterone, an inadequate response does not exclude AIS. Basal LH and testosterone may not be raised during early infancy. An LHRH stimulation test might be useful for evaluating cases of suspected AIS presenting in mid-childhood.


Assuntos
Síndrome de Resistência a Andrógenos/diagnóstico , Gonadotropinas Hipofisárias/biossíntese , Testosterona/biossíntese , Adolescente , Síndrome de Resistência a Andrógenos/fisiopatologia , Biomarcadores/sangue , Criança , Pré-Escolar , Gonadotropina Coriônica , Di-Hidrotestosterona/sangue , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/sangue , Hormônio Liberador de Gonadotropina , Gonadotropinas Hipofisárias/sangue , Humanos , Lactente , Hormônio Luteinizante/biossíntese , Hormônio Luteinizante/sangue , Masculino , Estudos Retrospectivos , Testosterona/sangue
15.
Endocrinology ; 139(4): 2015-24, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9528989

RESUMO

The presence of ovarian GnRH and/or compounds with GnRH-like activity was investigated in the goldfish ovary. Goldfish ovary was extracted using an acetone/HCl/ether mixture and was purified by Waters C-18 Sep-Pak columns (ovarian extract). The goldfish ovarian extract was found to 1) stimulate gonadotropin release and subunit messenger RNA production in the goldfish pituitary that was inhibited by a GnRH antagonist; 2) stimulate germinal vesicle breakdown in the prophase-I arrested follicle-enclosed goldfish oocytes in vitro, which was inhibited by a GnRH antagonist; 3) immunoreact with various GnRH antisera; and 4) bind to GnRH receptors in the goldfish pituitary and ovary as well as rat pituitary. Further purification with HPLC revealed the presence of two compounds with GnRH-like activity. One with identical chromatographic characteristics, amino acid composition, and primary structure to that of the salmon GnRH (sGnRH), and the other a novel compound with GnRH-like activity.


Assuntos
Carpa Dourada/metabolismo , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/análise , Ovário/química , Sequência de Aminoácidos , Animais , Cromatografia Líquida de Alta Pressão , Feminino , Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas Hipofisárias/biossíntese , Gonadotropinas Hipofisárias/metabolismo , Meiose/efeitos dos fármacos , Oócitos/citologia , Oócitos/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Receptores LHRH/metabolismo , Homologia de Sequência , Extratos de Tecidos/isolamento & purificação , Extratos de Tecidos/farmacologia
16.
Biol Reprod ; 57(5): 1145-54, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9369182

RESUMO

Gilthead seabream females undergo daily cycles of final oocyte maturation (FOM), ovulation, and spawning throughout their spawning season. FOM consists of lipid droplet and yolk granule coalescence, germinal vesicle (GV) migration, and GV breakdown. Plasma maturational gonadotropin (GtH-II) levels fluctuate throughout the day, reaching a peak at 8 h before spawning, when the GV is at the periphery of the oocyte. The preovulatory GtH-II surge is accompanied by an increase in the plasma levels of 17alpha,20beta-dihydroxy-4-pregnen-3-one and estradiol, while testosterone and 17alpha,20beta,21-trihydroxy-4-pregnen-3-one levels remain unchanged. Concurrent with the preovulatory GtH-II surge, there is an increase in pituitary GtH-II beta subunit mRNA levels followed by an increase in GtH-Ibeta mRNA levels. Gilthead seabream brain contains three different forms of GnRH: salmon (s)GnRH, seabream (sb)GnRH, and chicken (c)GnRH-II. All three GnRH-encoding mRNAs fluctuate throughout the day, reaching highest levels 8 h before spawning, concurrent with the preovulatory GtH-II surge. On the basis of these correlations and of the anatomical organization of the three GnRH systems, it is hypothesized that in the daily-spawning gilthead seabream females, preovulatory GtH-II secretion, and probably synthesis, are induced by a surge of sbGnRH secretion. The involvement of the other two GnRH forms, sGnRH and cGnRH-II, in the control of ovulation and spawning is presumed, on the basis of the elevation of their mRNA levels at the time of the preovulatory GtH-II secretion and spawning.


Assuntos
Fase Folicular/fisiologia , Hormônio Liberador de Gonadotropina/biossíntese , Gonadotropinas Hipofisárias/biossíntese , Gonadotropinas/sangue , Perciformes/fisiologia , RNA Mensageiro/biossíntese , Esteroides/sangue , Animais , Autorradiografia , Estradiol/sangue , Feminino , Oócitos/crescimento & desenvolvimento , Ovário/anatomia & histologia , Ovário/citologia , Perciformes/sangue , Testosterona/sangue
17.
Gen Comp Endocrinol ; 104(1): 103-15, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8921361

RESUMO

To analyze the multihormonal control mechanisms of GTH secretion in the eel, primary culture of pituitary cells from control or estradiol-treated female silver eels, a treatment known to stimulate in vivo GTH synthesis, was developed. Dispersed eel pituitary cells obtained by enzymatic (trypsin/DNAse) and mechanical dispersion were cultured in Earles M199, at 18 degrees. Immunoreactive GTH (ir-GTH) cells were characterized by the immunoperoxidase method, using antiserum to carp GTH beta subunit. Ir-GTH cells from control silver eels were small and represented 14% of the dispersed pituitary cells. In contrast, ir-GTH cells from estradiol-treated eels were larger (cell area x 2.5) and represented a higher proportion (21%) of the pituitary cells. Intracellular and medium contents of GTH were measured by radioimmunoassay for the GTH beta subunit. In vivo estradiol-treatment increased more than 100-fold the GTH content of cell cultures. GTH release, studied over 1 to 4 hr, was undetectable in cultures from normal eels. In contrast, GTH release was low (less than 2% of cell content) but measurable in cultures from estradiol-treated eels. Subsequent experiments examined effects of various secretagogues on GTH release from primary cultures of pituitary cells from estradiol-pretreated eels. GTH release was significantly increased (x1.5 to x3 basal release) by 10(-6) M GnRH-A as well as by both native GnRHs in the eel (mammalian GnRH, mGnRH, and chicken GnRH-II, cGnRH-II), at the same concentration. Lower GnRH concentrations had no significant effect, indicating a low sensitivity of gonadotrophs to GnRH, likely to be related to their immature state at the silver stage. The similar efficacy of mGnRH and cGnRH-II suggested that the pituitary GnRH receptor had a low specificity toward various molecular forms, in the eel as in the other nonmammalian species. The protein kinase C (PKC) activator (phorbol ester: PMA) also stimulated GTH secretion, with a maximal effect at 10(-8) M, indicating that the PKC pathway was functional. In contrast, a depolarizing agent (50 mM KCl) had no significant effect on GTH release, suggesting lack of a functional voltagesensitive calcium channel (VSCC) secretory pathway. Perifusion experiments on whole pituitary confirmed the lack of effect of KCl on gonadotrophs from E2-pretreated eels and indicated that an additional in vivo treatment with GnRH agonist and dopamine antagonist could induce the differentiation of a functional VSCC pathway. These characteristics of the transduction mechanisms may be related to the immature state of the eel gonadotrophs at the silver stage.


Assuntos
Anguilla/metabolismo , Estradiol/farmacologia , Gonadotropinas Hipofisárias/biossíntese , Hipófise/metabolismo , Animais , Células Cultivadas , Feminino , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Imuno-Histoquímica , Perfusão , Ésteres de Forbol/farmacologia , Hipófise/citologia , Hipófise/efeitos dos fármacos , Cloreto de Potássio/farmacologia , Radioimunoensaio
18.
Gen Comp Endocrinol ; 102(3): 327-41, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8804563

RESUMO

In the goldfish, it has been proposed that gonadotropin (GTH) release induced by GTH-releasing hormone (GnRH) involves Ca2+ entry through voltage-sensitive Ca2+ channels (VSCC), protein kinase C (PKC) activation, and arachidonic acid (AA) metabolism, but not cyclic AMP (cAMP) action. However, cAMP appears to mediate GnRH action in other teleosts. In this study, the relative importance of PKC and cAMP in mediating GnRH action in goldfish was studied using primary cultures of dispersed pituitary cells. Consistent with an involvement of PKC in GnRH action, the GTH responses to the PKC activating tetradecanoyl phorbol acetate (TPA), salmon (s)GnRH, and chicken (c)GnRH-II were inhibited by two selective PKC inhibitors, calphostin C, and staurosporine. Furthermore, GTH release responses induced by sGnRH or cGnRH-II were not additive to responses stimulated by the PKC-activating diglyceride DiC8, in either long-term static incubation or acute perifusion experiments. In static incubation studies, the GTH responses to sGnRH and DiC8 were potentiated by the VSCC agonist Bay K 8644, suggesting that VSCC participates in both PKC and GnRH action. Concentrations of K+ < 100 mM did not elicit GTH secretion when tested alone, but were effective in stimulating GTH release in the presence of subthreshold doses of DiC8 or TPA. This suggests that minimal activation of PKC greatly enhances the effectiveness of Ca2+ influx to increase GTH secretion. Taken together, these results indicate that PKC is an important mediator of GnRH-induced, VSCC-dependent GTH release. In contrast to the involvement of PKG, cAMP-dependent mechanisms showed no evidence of direct participation in GnRH-induced GTH release in goldfish. In static incubation studies, the GTH responses to sGnRH and cGnRH-II were not affected by H89, a cAMP-dependent protein kinase (PKA) inhibitor. Furthermore, the GTH release stimulated by cAMP was additive to the response to sGnRH, cGnRH-II, DiC8, TPA, or AA. However, compared to the response to forskolin or TPA alone, combinations of forskolin and TPA resulted in a potentiated increase in GTH release. The acute GTH response to forskolin was also enhanced by DiC8. Thus, cAMP-dependent mechanisms may constitute an independent pathway that interacts positively with GnRH-dependent mechanisms in the regulation of GTH release.


Assuntos
AMP Cíclico/metabolismo , Carpa Dourada/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas Hipofisárias/metabolismo , Hipófise/metabolismo , Proteína Quinase C/metabolismo , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Alcaloides/farmacologia , Animais , Ácido Araquidônico/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Células Cultivadas , Galinhas , Colforsina/farmacologia , AMP Cíclico/farmacologia , Diglicerídeos/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas Hipofisárias/biossíntese , Masculino , Microesferas , Naftalenos/farmacologia , Hipófise/citologia , Potássio/farmacologia , Proteína Quinase C/efeitos dos fármacos , Salmão , Transdução de Sinais , Estaurosporina , Acetato de Tetradecanoilforbol/farmacologia
19.
Mol Endocrinol ; 10(3): 217-29, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8833651

RESUMO

The orphan nuclear receptor steroidogenic factor-1 (SF-1) regulates the expression of several genes involved in the reproductive function and development of the adrenal, the gonads, and the pituitary gonadotropes. It also confers the gonadotrope-specific expression of the glycoprotein hormone a subunit gene by the binding to a gonadotrope-specific element (GSE). In this study, we have shown that SF-1 transactivates the salmon gonadotropin II beta subunit (sGTHII beta) gene expression. SF-1 alone offered a slight but significant enhancement on sGTHII beta promoter activity (7.2 +/- 0.6 fold). However, it stimulated sGTHII beta gene expression dramatically (127 +/- 37 fold) when combined with the estrogen receptor (ER). This synergistic interaction was specific for sGTHII beta promoter as well as for both SF-1 and ER and was estradiol-dose dependent. 5'-Deletion studies of the sGTHII beta promoter identified two putative SF-1 binding sites (GSE) and one previously identified proximal estrogen-responsive element (pERE) at -274 bp involved in this activation. The two GSE sequences located at -354 bp (sGSE(3) and -162 bp (sGSE(2) upstream of the transcription site, although imperfect as compared with the consensus GSE, bound specifically to the in vitro-translated mouse SF-1 protein. 5'-Deletion studies, competition experiments, and site-directed mutagenesis showed that binding to pERE and GSE(2) were necessary for the SF-1/ER synergistic effect. These studies suggest that the synergistic interaction of SF-1 and ER, possibly through cooperative binding or protein-protein interaction, is essential in conferring a cell type-specific expression of the GTHII beta subunit gene.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Gonadotropinas Hipofisárias/genética , Receptores de Estradiol/fisiologia , Salmão/genética , Fatores de Transcrição/fisiologia , Animais , Sequência Consenso , Fatores de Transcrição Fushi Tarazu , Genes , Gonadotropinas Hipofisárias/biossíntese , Células HeLa , Proteínas de Homeodomínio , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Especificidade de Órgãos , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares , Proteínas Recombinantes de Fusão/biossíntese , Sequências Reguladoras de Ácido Nucleico , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Fator Esteroidogênico 1 , Estimulação Química , Transcrição Gênica , Transfecção
20.
Mol Cell Biol ; 15(7): 3531-9, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7791760

RESUMO

Gonadotropin-releasing hormone (GnRH) interacts with a G protein-coupled receptor and increases the transcription of the glycoprotein hormone alpha-subunit gene. We have explored the possibility that mitogen-activated protein kinase (MAPK) plays a role in mediating GnRH effects on transcription. Activation of the MAPK cascade by an expression vector for a constitutively active form of the Raf-1 kinase led to stimulation of the alpha-subunit promoter in a concentration-dependent manner. GnRH treatment was found to increase the phosphorylation of tyrosine residues of MAPK and to increase MAPK activity, as determined by an immune complex kinase assay. A reporter gene assay using the MAPK-responsive, carboxy-terminal domain of the Elk1 transcription factor was also consistent with GnRH-induced activation of MAPK. Interference with the MAPK pathway by expression vectors for kinase-defective MAPKs or vectors encoding MAPK phosphatases reduced the transcription-stimulating effects of GnRH. The DNA sequences which are required for responses to GnRH include an Ets factor-binding site. An expression vector for a dominant negative form of Ets-2 was able to reduce GnRH effects on expression of the alpha-subunit gene. These findings provide evidence that GnRH treatment leads to activation of the MAPK cascade in gonadotropes and that activation of MAPK contributes to stimulation of the alpha-subunit promoter. It is likely that an Ets factor serves as a downstream transcriptional effector of MAPK in this system.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas Hipofisárias/biossíntese , Hipófise/efeitos dos fármacos , Transdução de Sinais , Animais , Sítios de Ligação , Western Blotting , Linhagem Celular , Células Cultivadas , Regulação da Expressão Gênica , Genes Reporter , Gonadotropinas Hipofisárias/genética , Proteínas Imediatamente Precoces/biossíntese , Camundongos , Hipófise/citologia , Hipófise/metabolismo , Testes de Precipitina , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Fosfatases/biossíntese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-raf , Proteínas Oncogênicas de Retroviridae/metabolismo , Transcrição Gênica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA