Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neuroendocrinol ; 25(5): 488-96, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23331967

RESUMO

Tonic gonadotrophin secretion throughout the menstrual cycle is regulated by the negative-feedback actions of ovarian oestradiol (E2) and progesterone. Although kisspeptin neurones in the arcuate nucleus (ARC) of the hypothalamus appear to play a major role in mediating these feedback actions of the steroids in nonprimate species, this issue has been less well studied in the monkey. In the present study, we used immunohistochemistry and in situ hybridisation to examine kisspeptin and KISS1 expression, respectively, in the mediobasal hypothalamus (MBH) of adult ovariectomised (OVX) rhesus monkeys. We also examined kisspeptin expression in the MBH of ovarian intact females, and the effect of E2, progesterone and E2 + progesterone replacement on KISS1 expression in OVX animals. Kisspeptin or KISS1 expressing neurones and pronounced kisspeptin fibres were readily identified throughout the ARC of ovariectomised monkeys but, on the other hand, in intact animals, kisspeptin cell bodies were small in size and number and only fine fibres were observed. Replacement of OVX monkeys with physiological levels of E2, either alone or with luteal phase levels of progesterone, abolished KISS1 expression in the ARC. Interestingly, progesterone replacement alone for 14 days also resulted in a significant down-regulation of KISS1 expression. These findings support the view that, in primates, as in rodents and sheep, kisspeptin signalling in ARC neurones appears to play an important role in mediating the negative-feedback action of E2 on gonadotrophin secretion, and also indicate the need to study further their regulation by progesterone.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Kisspeptinas/metabolismo , Neurônios/metabolismo , Ovário/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Estradiol/administração & dosagem , Feminino , Hipotálamo/metabolismo , Macaca mulatta , Monoaminoxidase/genética , Ovariectomia , Progesterona/administração & dosagem , RNA Mensageiro/genética
2.
Endocrinology ; 152(8): 3182-91, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21628385

RESUMO

Previously, we reported that 1 nM 17ß-estradiol (E(2)) induces a rapid action, which is, in part, mediated through the G protein-coupled receptor GPR30 in primate GnRH neurons. Because it has been reported that the diphenylacrylamide compound, STX, causes estrogenic action in the mouse and guinea pig hypothalamus, the present study examined effects of STX in primate GnRH neurons and whether there is an action independent of GPR30. Results are summarized as follows. STX (10 nM) exposure increased 1) the oscillation frequency of intracellular calcium concentration ([Ca(2+)](i)), 2) the percentage of cells stimulated, and 3) the synchronization frequency of [Ca(2+)](i) oscillations. STX (10-100 nM) also stimulated GnRH release. The effects of STX on both [Ca(2+)](i) oscillations and GnRH release were similar to those caused by E(2) (1 nM), although with less magnitude. STX (10 nM)-induced changes in [Ca(2+)](i) oscillations were not altered by GPR30 small interfering RNA transfection, indicating that STX-sensitive receptors differ from GPR30. Finally, a higher dose of E(2) (10 nM) induced a larger change in [Ca(2+)](i) oscillations than that with a smaller dose of E(2) (1 nM), and the effects of 10 nM E(2) were reduced but not completely blocked by GPR30 small interfering RNA transfection, indicating that the effects of 10 nM E(2) in primate GnRH neurons are mediated by multiple membrane receptors, including GPR30 and STX-sensitive receptors. Collectively, the rapid action of E(2) mediated through GPR30 differs from that mediated through STX-sensitive receptors. The molecular structure of the STX-sensitive receptor remains to be identified.


Assuntos
Acrilamidas/farmacologia , Cálcio/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/efeitos dos fármacos , Animais , Estradiol/análogos & derivados , Estradiol/farmacologia , Fulvestranto , Macaca mulatta , Neurônios/metabolismo , Receptores de Estrogênio , Receptores Acoplados a Proteínas G/fisiologia
3.
Steroids ; 76(9): 861-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21354432

RESUMO

Estrogens play a pivotal role in the control of female reproductive function. Recent studies using primate GnRH neurons derived from embryonic nasal placode indicate that 17ß-estradiol (E(2)) causes a rapid stimulatory action. E(2) (1nM) stimulates firing activity and intracellular calcium ([Ca(2+)](i)) oscillations of primate GnRH neurons within a few min. E(2) also stimulates GnRH release within 10min. However, the classical estrogen receptors, ERα and ERß, do not appear to play a role in E(2)-induced [Ca(2+)](i) oscillations or GnRH release, as the estrogen receptor antagonist, ICI 182,780, failed to block these responses. Rather, this rapid E(2) action is, at least in part, mediated by a G-protein coupled receptor GPR30. In the present study we further investigate the role of ERα and ERß in the rapid action of E(2) by knocking down cellular ERα and ERß by transfection of GnRH neurons with specific siRNA for rhesus monkey ERα and ERß. Results indicate that cellular knockdown of ERα and ERß failed to block the E(2)-induced changes in [Ca(2+)](i) oscillations. It is concluded that neither ERα nor ERß is required for the rapid action of E(2) in primate GnRH neurons.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Estrogênios/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Condutos Olfatórios/metabolismo , Animais , Sinalização do Cálcio , Estradiol/fisiologia , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Estrogênios/fisiologia , Macaca mulatta , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Condutos Olfatórios/citologia , Condutos Olfatórios/embriologia , Interferência de RNA , Técnicas de Cultura de Tecidos
4.
J Neuroendocrinol ; 22(7): 630-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20456608

RESUMO

Since Ernst Knobil proposed the concept of the gonadotrophin-releasing hormone (GnRH) pulse-generator in the monkey hypothalamus three decades ago, we have made significant progress in this research area with cellular and molecular approaches. First, an increase in pulsatile GnRH release triggers the onset of puberty. However, the question of what triggers the pubertal increase in GnRH is still unclear. GnRH neurones are already mature before puberty but GnRH release is suppressed by a tonic GABA inhibition. Our recent work indicates that blocking endogenous GABA inhibition with the GABA(A) receptor blocker, bicuculline, dramatically increases kisspeptin release, which plays an important role in the pubertal increase in GnRH release. Thus, an interplay between the GABA, kisspeptin, and GnRH neuronal systems appears to trigger puberty. Second, cultured GnRH neurones derived from the olfactory placode of monkey embryos exhibit synchronised intracellular calcium, [Ca(2+)](i), oscillations and release GnRH in pulses at approximately 60-min intervals after 14 days in vitro (div). During the first 14 div, GnRH neurones undergo maturational changes from no [Ca(2+)](i) oscillations and little GnRH release to the fully functional state. Recent work also shows GnRH mRNA expression increases during in vitro maturation. This mRNA increase coincides with significant demethylation of a CpG island in the GnRH 5'-promoter region. This suggests that epigenetic differentiation occurs during GnRH neuronal maturation. Third, oestradiol causes rapid, direct, excitatory action in GnRH neurones and this action of oestradiol appears to be mediated through a membrane receptor, such as G-protein coupled receptor 30.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/fisiologia , Primatas/anatomia & histologia , Primatas/metabolismo , Animais , Epigênese Genética , Estradiol/metabolismo , Hormônio Liberador de Gonadotropina/genética , Neurônios/citologia , Puberdade/fisiologia , Maturidade Sexual/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Ácido gama-Aminobutírico/metabolismo
5.
J Neuroendocrinol ; 21(4): 316-21, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19207808

RESUMO

Previously, we have shown that 17beta-oestradiol (E(2)) induces an increase in firing activity and modifies the pattern of intracellular calcium ([Ca(2+)](i)) oscillations with a latency < 1 min in primate luteinising hormone-releasing hormone (LHRH) neurones. A recent study also indicates that E(2), the nuclear membrane impermeable oestrogen, oestrogen-dendrimer conjugate, and the plasma membrane impermeable oestrogen, E(2)-BSA conjugate, all similarly stimulated LHRH release within 10 min of exposure in primate LHRH neurones, indicating that the rapid action of E(2) is caused by membrane signalling. The results from a series of studies further suggest that the rapid action of E(2) in primate LHRH neurones appears to be mediated by GPR30. Although the oestrogen receptor antagonist, ICI 182, 780, neither blocked the E(2)-induced LHRH release nor the E(2)-induced changes in [Ca(2+)](i) oscillations, E(2) application to cells treated with pertussis toxin failed to result in these changes in primate LHRH neurones. Moreover, knockdown of GPR30 in primate LHRH neurones by transfection with human small interference RNA for GPR30 completely abrogated the E(2)-induced changes in [Ca(2+)](i) oscillations, whereas transfection with control siRNA did not. Finally, the GPR30 agonist, G1, resulted in changes in [Ca(2+)](i) oscillations similar to those observed with E(2). In this review, we discuss the possible role of G-protein coupled receptors in the rapid action of oestrogen in neuronal cells.


Assuntos
Estrogênios/metabolismo , Hormônio Luteinizante/metabolismo , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Cálcio/metabolismo , Membrana Celular/fisiologia , Estradiol/farmacologia , Receptor beta de Estrogênio/metabolismo , Estrogênios/farmacologia , Neurônios/efeitos dos fármacos , Periodicidade , Fatores de Tempo
6.
J Neurophysiol ; 88(3): 1559-67, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12205175

RESUMO

Periodic release of luteinizing hormone-releasing hormone (LHRH) from the hypothalamus is essential for normal reproductive function. Pulsatile LHRH release appears to result from the synchronous activity of LHRH neurons. However, how the activity of these neurons is synchronized to release LHRH peptide in a pulsatile manner is unclear. Because there is little evidence of physical coupling among LHRH neurons in the hypothalamus, we hypothesized that the activity of LHRH neurons might be coordinated by indirect intercellular communication via intermediary (nonneural) cells rather than direct interneural coupling. In this study, we used an in vitro preparation of LHRH neurons derived from the olfactory placode of monkey embryos to assess whether nonneuronal cells, play a role in coordinating LHRH neuronal activity. We found that cultured LHRH neurons and nonneuronal cells both exhibit spontaneous oscillations in the concentration of intracellular Ca(2+) ([Ca(2+)](i)) at similar frequencies. Moreover, [Ca(2+)](i) oscillations in both types of cell were periodically synchronized. Synchronized [Ca(2+)](i) oscillations spread as intercellular Ca(2+) waves across fields of cells that included LHRH neurons and nonneuronal cells, although waves spread at a higher velocity among LHRH neurons. These results suggest that LHRH neurons and nonneuronal cells are functionally integrated and that nonneuronal cells could be involved in synchronizing the activity of the LHRH neurosecretory network.


Assuntos
Cálcio/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Animais , Células/metabolismo , Técnicas de Cultura , Ectoderma/citologia , Ectoderma/metabolismo , Membranas Intracelulares/metabolismo , Macaca mulatta/embriologia , Condutos Olfatórios/citologia , Condutos Olfatórios/embriologia , Oscilometria , Concentração Osmolar , Fatores de Tempo
7.
Endocrinology ; 140(11): 5257-66, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10537156

RESUMO

In order to test the hypothesis that GABA is an inhibitory neurotransmitter restricting the release of LHRH before puberty, we examined the effects of pulsatile infusion of the GABA(A) receptor blocker, bicuculline, on the timing of puberty. Eleven female monkeys at 14-15 months of age were implanted with a stainless steel cannula into the base of the third ventricle above the median eminence. Five monkeys received bicuculline infusion every 2 h at a dose of 1 microM with a gradual increase to 100 microM in 10 microl using a portable infusion pump. The remaining 6 monkeys received similar infusions of saline. An additional 11 colony monkeys without cannula implantation were used for controls. Results indicate that bicuculline infusion advances the timing of puberty. The age of menarche (17.8+/-0.5 months) in the bicuculline infusion animals was significantly earlier than that in the saline controls (28.2+/-2.3, P < 0.001) as well as in colony controls (30.6+/-0.9, P < 0.001). The age of first ovulation (30.5+/-3.3 months) in bicuculline-treated animals was much younger (P < 0.001) than that in both controls (44.8+/-1.8 and 44.7+/-1.2, respectively). Bicuculline also accelerated the growth curve. These results suggest that the reduction of tonic GABA inhibition of LHRH neurons advances the onset of puberty.


Assuntos
Bicuculina/farmacologia , Antagonistas GABAérgicos/farmacologia , Antagonistas de Receptores de GABA-A , Macaca mulatta/fisiologia , Maturidade Sexual/efeitos dos fármacos , Envelhecimento , Animais , Bicuculina/administração & dosagem , Peso Corporal , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante/metabolismo , Antagonistas GABAérgicos/administração & dosagem , Hormônio do Crescimento/sangue , Hormônio Luteinizante/sangue , Hormônio Luteinizante/metabolismo , Eminência Mediana/efeitos dos fármacos , Menstruação/fisiologia , Ovulação/fisiologia , Periodicidade , Progesterona/sangue
8.
J Neurosci ; 19(14): 5898-909, 1999 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10407029

RESUMO

To understand the mechanism of pulsatile luteinizing hormone-releasing hormone (LHRH) release, we examined whether cultured LHRH neurons exhibit spontaneous intracellular Ca(2+) ([Ca(2+)](i)) signaling. The olfactory placode and the ventral migratory pathway of LHRH neurons from rhesus monkey embryos at embryonic ages 35-37 were dissected out and cultured on glass coverslips. Two to five weeks later, cultured cells were labeled with fura-2 and examined for [Ca(2+)](i) signaling by recording changes in [Ca(2+)](i) every 10 sec for 30-175 min. Cells were fixed and immunostained for LHRH and neuron-specific enolase. In 20 cultures, 572 LHRH-positive cells exhibited [Ca(2+)](i) oscillations at an interpulse interval (IPI) of 8.2 +/- 0.7 min and a duration of 88.8 +/- 2.9 sec. LHRH-negative neurons in culture exhibited only occasional [Ca(2+)](i) oscillations. In 17 of 20 cultures with LHRH-positive cells, [Ca(2+)](i) oscillations occurred synchronously in 50-100% of the individual cells, whereas [Ca(2+)](i) oscillations in cells in the remaining three cultures did not synchronize. Strikingly, in 12 of 17 cultures the synchronization of [Ca(2+)](i) oscillations repeatedly occurred in complete unison at 52.8 +/- 3.0 min intervals, which is similar to the period observed for LHRH release, whereas in 5 of 17 cultures the less tight synchronization of [Ca(2+)](i) oscillations repeatedly occurred at 23.4 +/- 4.6 min intervals. IPI of [Ca(2+)](i) oscillations in cells with tight synchronization and less tight synchronization did not differ from IPI in cells without synchronization. The results indicate that LHRH neurons derived from the monkey olfactory placode possess an endogenous mechanism for synchronization of [Ca(2+)](i) oscillations. Whether synchronization of [Ca(2+)](i) oscillations relates to neurosecretion remains to be investigated.


Assuntos
Cálcio/metabolismo , Hormônio Liberador de Gonadotropina/fisiologia , Neurônios/fisiologia , Condutos Olfatórios/fisiologia , Ciclos de Atividade , Animais , Células Cultivadas , Embrião de Mamíferos , Feto , Imuno-Histoquímica , Macaca mulatta , Neurônios/citologia , Condutos Olfatórios/embriologia , Oscilometria , Transdução de Sinais
9.
Endocrinology ; 140(3): 1432-41, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10067872

RESUMO

To study the mechanism of LH-releasing hormone (LHRH) pulse generation, the olfactory pit/placode and the migratory pathway of LHRH neurons from monkey embryos at embryonic age 35-37 were dissected out, under the microscope, and cultured on plastic coverslips coated with collagen in a defined medium for 2-5 weeks. First, we examined whether cultured neurons release the decapeptide into media. It was found that LHRH cells release LHRH in a pulsatile manner at approximately 50-min intervals. Further, LHRH release was stimulated by depolarization with high K+ and the Na+ channel opener, veratridine. However, whereas the Na+ channel blocker, tetrodotoxin suppressed the effects of veratridine, tetrodotoxin did not alter the effects of high K+. Subsequently, the role of extracellular and intracellular Ca2+ in LHRH release was examined. The results are summarized as follows: 1) exposing the cells to a low Ca2+ (20 nM) buffer solution suppressed LHRH release, whereas exposure to a normal Ca2+ solution (1.25 mM) maintained pulsatile LHRH release; 2) LHRH release from cultured LHRH cells was stimulated by the voltage-sensitive L-type Ca2+ channel agonist, Bay K 8644 (10 microM), whereas it was suppressed by the L-type Ca2+ channel blocker, nifedipine (1 microM), but not by the N-type channel blocker, omega-conotoxin GVIA (1 microM); 3) the intracellular Ca2+ stimulant, ryanodine (1 microM), stimulated LHRH release, whereas the intracellular Ca2+ transporting adenosine triphosphatase antagonist, thapsigargin (1 and 10 microM), did not yield consistent results; and 4) carbonyl cyanide p-trifluoromethoxyphenyl-hydrazone (1 microM), a mitochondrial Ca2+ mobilizer, stimulated LHRH release, whereas ruthenium red, a mitochondrial Ca2+ uptake inhibitor, did not induce consistent results. These results indicate that: 1) the presence of extracellular Ca2+ is essential for LHRH neurosecretion; 2) Ca2+ enters the cell via L-type channels but not N-type channels; and 3) mobilization of intracellular Ca2+ from inositol 1,4,5-triphosphate-sensitive stores, as well as mitochondrial stores, seem to contribute to LHRH release in these cells.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Condutos Olfatórios/embriologia , Animais , Cálcio/metabolismo , Canais de Cálcio/efeitos dos fármacos , Células Cultivadas , Embrião de Mamíferos/citologia , Macaca mulatta , Condutos Olfatórios/citologia , Taxa Secretória
10.
J Neurosci ; 16(8): 2563-73, 1996 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-8786432

RESUMO

To investigate further the role of GABA in the onset of puberty, this study examines whether glutamic acid decarboxylase (GAD), the catalytic enzyme for GABA synthesis, is involved in the suppression of luteinizing hormone releasing hormone (LHRH) before puberty in rhesus monkeys. First, both GAD67 and GAD65 mRNAs were detectable by reverse transcription-PCR analysis in the preoptic area, medio-basal hypothalamus, posterior hypothalamic area, and hippocampus of the monkey brain. Second, effects of antisense oligodeoxynucleotides (D-oligos) for GAD67 and GAD65 mRNAs on LHRH release were examined in conscious female rhesus monkeys at the prepubertal stage using a push-pull perfusion method. The GAD67 or GAD65 antisense D-oligos or scrambled D-oligos were infused directly into the stalk-median eminence. Both the GAD67 and the GAD65 antisense D-oligos induced a large and prompt increase in LHRH release, whereas the scrambled D-oligos did not induce any significant effect. The results suggest that the removal of GABA inhibition by interfering with GAD synthesis is effective in increasing LHRH release in prepubertal monkeys. Third, the specificity of the antisense D-oligos on GAD levels was examined by incubating basal hypothalami with D-oligos in vitro and subsequent Western blot analysis. The antisense D-oligos consistently decreased the proteins GAD67 and GAD65 compared with respective control D-oligos. We conclude that the decrease of tonic GABAergic inhibition and maturational changes in GAD synthesis may be critical factors for the onset of puberty in nonhuman primates.


Assuntos
Glutamato Descarboxilase/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Animais , Sequência de Bases , Feminino , Macaca mulatta , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...