Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Mol Cell Endocrinol ; 203(1-2): 25-32, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12782400

RESUMO

The number of pituitary cells, their size, hormonal content and release and response to external cues varies between day and night and during the estrus cycle. Previous studies have demonstrated that pituitary cells proliferate rhythmically and that estradiol (E(2)) is a mitogen of alpha T3 cells. We, therefore, studied the effect of gonadotropin releasing hormone (GnRH) and E(2), on the cell cycle in primary cultures of mouse pituitary cells and in the gonadotroph cell line L beta T2. We found that GnRH and E(2) modulate the cell cycle in a time dependent manner and induce proliferation in cultures of mouse pituitary and L beta T2 cells. GnRH induces proliferation in cells isolated in the morning of the estrus day and increases the number of cells in G2 stage when isolated in noon and evening. However, the transition into the G1 stage is enabled only by co-addition of E(2) and GnRH. GnRH stimulates LH release from L beta T2 cells after 2 days via exocytosis while after 4 days in culture, the increase in LH release may be accounted for by the increase in cell number. E(2) enhanced the GnRH response after 2 days, and abolished it after 4 days in culture. Furthermore, E(2) has no effect on LH release and cell number after 2 days in culture, however, after 4 days in culture, E(2) had no effect on the total amount of LH released but inhibited LH release per cell due to increase in cell number. Our results show that GnRH and E(2) function to shorten the cell cycle and regulate the cell number of each stage of the cell cycle. The effect of GnRH and E(2) on the cell cycle is dependent on the circadian time. This mechanism may serve to modulate the size and function of the pituitary cell population and consequently the function of pituitary gonadotrophs regulating the surge of LH release before ovulation.


Assuntos
Ciclo Celular/efeitos dos fármacos , Estradiol/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Hipófise/citologia , Animais , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Ritmo Circadiano , Sinergismo Farmacológico , Fase G1 , Hormônio Luteinizante/metabolismo , Camundongos , Periodicidade , Fatores de Tempo
3.
Mol Cell Endocrinol ; 189(1-2): 125-34, 2002 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-12039071

RESUMO

Exposure of tilapia pituitary cells in culture to salmon gonadotropin-releasing hormone (sGnRH; 0.01-100 nM) elevated the phosphorylated extracellular signal-regulated kinase (pERK) levels. sGnRH also elevated the alpha, FSHbeta and LHbeta subunit mRNA levels. The phorbol ester, 1-O-tetradecanoyl phorbol-13-acetate (TPA; 12.5 nM) increased pERK levels, whereas protein kinase C (PKC) depletion or inhibition by GF109203X (GF; 0.01-10 microM) suppressed GnRH-activated ERKs. GF too abated the GnRH-induced alpha and LHbeta mRNA levels, but had no effect on those of FSHbeta. Forskolin (0.001-100 microM) activated ERK, while inhibition of protein kinase A (PKA) by H89 (0.01-10 microM) suppressed pERK levels and all GnRH-stimulated gonadotropin subunit transcripts. Exposure of cells to the mitogen-activated protein kinase kinase (MAPK kinase; MEK) inhibitor (PD98059; PD 10, 25 and 50 microM) completely blocked GnRH-induced increase in ERKs activation. Furthermore, PD suppressed the alpha and LHbeta mRNA responses to GnRH, but had no effect on FSHbeta mRNA levels. It is suggested that in tilapia the differential regulation of gonadotropin subunit gene expression by GnRH results from a divergent recruitment of signal transduction pathways, activated upon GnRH binding; PKC-ERK cascade is involved in elevating alpha and LHbeta mRNAs, whereas induction of FSHbeta transcript is ERK-independent and is under direct cAMP-PKA regulation or through other MAPK cascades.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas Hipofisárias/genética , Hipófise/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tilápia/fisiologia , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Gonadotropinas Hipofisárias/metabolismo , Indóis/farmacologia , Masculino , Maleimidas/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Hipófise/citologia , Hipófise/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Subunidades Proteicas , Salmão/metabolismo , Transdução de Sinais/fisiologia
4.
Arch Med Res ; 32(6): 499-509, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11750725

RESUMO

The hypothalamic gonadotropin-releasing hormone (GnRH) is a key regulator of the reproductive system, triggering the synthesis and release of LH and FSH in the pituitary. GnRH transmits its signal via two specific serpentine receptors that belong to the large group of G-protein coupled receptors (GPCRs). Here we review the intracellular signaling pathways mediated by the GnRH receptor (GnRHR). In pituitary-derived alpha T3-1 cells, a widely used model for GnRH action, GnRHR signaling includes activation of mitogen-activated protein kinase (MAPK) cascades, which provide an important link for the transmission of signals from the cell surface to the nucleus and play a role in the regulation of gonadotropin transcription. Activation of ERK--one of the MAPK cascades--by GnRH in these cells depends mainly on the phosphorylation of Raf1 by PKC, supported by a pathway involving c-Src, dynamin, and Ras. On the other hand, the activation of JNK, another MAPK cascade, involves PKC, c-Src, CDC42/Rac1, and probably MEKK1. The GnRHR is also expressed in non-pituitary cells and was found to be involved in the inhibition of cell proliferation in certain cells. Therefore, GnRHR represents a potential target for GnRH-analogs used for cancer treatment. Interestingly, the signaling mechanism of the GnRHR in other cell types significantly differs from that in pituitary cells. Studies conducted in GnRHR-expressing COS7 cells have shown that GnRHR transmits its signals mainly via Gi, EGF receptor, c-Src, and is not dependent on PKC. Understanding the signaling mechanisms elicited by GnRHR can shed light on the mechanism of action of GnRH in pituitary and extra-pituitary tissues.


Assuntos
Sinalização do Cálcio/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Receptores LHRH/fisiologia , Animais , Antineoplásicos Hormonais/farmacologia , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/patologia , Células COS , Sinalização do Cálcio/efeitos dos fármacos , Chlorocebus aethiops , Ativação Enzimática , Feminino , Proteínas de Ligação ao GTP/fisiologia , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Sistema Hipotálamo-Hipofisário/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Modelos Biológicos , Fosforilação , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/fisiologia , Neoplasias Hipofisárias/patologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Proteína Quinase C/fisiologia , Processamento de Proteína Pós-Traducional , Receptores LHRH/efeitos dos fármacos , Transcrição Gênica , Células Tumorais Cultivadas/efeitos dos fármacos , Vertebrados/fisiologia
5.
Med Res Rev ; 21(5): 472-83, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11579443

RESUMO

G-protein-coupled receptors (GPCRs) are a large and functionally diverse protein superfamily, which form a seven transmembrane (TM) helices bundle with alternating extra-cellular and intracellular loops. GPCRs are considered to be one of the most important groups of drug targets because they are involved in a broad range of body functions and processes and are related to major diseases. In this paper we present a new technology, named PREDICT, for modeling the 3D structure of any GPCR from its amino acid sequence. This approach takes into account both internal protein properties (i.e., the amino acid sequence) and the properties of the membrane environment. Unlike competing approaches, the new technology does not rely on the single known structure of rhodopsin, and is thus capable of predicting novel GPCR conformations. We demonstrate the capabilities of PREDICT in reproducing the known experimental structure of rhodopsin. In principle, PREDICT-generated models offer new opportunities for structure-based drug discovery towards GPCR targets.


Assuntos
Proteínas de Ligação ao GTP/química , Modelos Estruturais , Receptores de Superfície Celular/química , Sequência de Aminoácidos , Animais , Desenho de Fármacos , Humanos , Conformação Proteica
6.
Chronobiol Int ; 18(3): 399-412, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11475411

RESUMO

The possibility that the 24h rhythm output is the composite expression of ultradian oscillators of varying periodicities was examined by assessing the effect of external continuously or pulsed (20-minute) Gonadotropin-releasing hormone (GnRH) infusions on in vitro luteinizing hormone (LH) release patterns from female mouse pituitaries during 38h study spans. Applying stepwise analyses (spectral, cosine fit, best-fit curve, and peak detection analyses) revealed the waveform shape of LH release output patterns over time is composed of several ultradian oscillations of different periods. The results further substantiated previous observations indicating the pituitary functions as an autonomous clock. The GnRH oscillator functions as a pulse generator and amplitude regulator, but it is not the oscillator that drives the ultradian LH release rhythms. At different stages of the estrus cycle, the effect of GnRH on the expression of ultradian periodicities varies, resulting in the modification of their amplitudes but not their periods. The functional output from the system of ultradian oscillators may superimpose a "circadian or infradian phenotype" on the observed secretion pattern. An "amplitude control" hypothesis is proposed: The temporal pattern of LH release is governed by several oscillators that function in conjunction with one another and are regulated by an amplitude-controlled mechanism. Simulated models show that such a mechanism results in better adaptive response to environmental requirements than does a single circadian oscillator.


Assuntos
Ritmo Circadiano , Hormônio Luteinizante/metabolismo , Animais , Feminino , Análise de Fourier , Hormônio Liberador de Gonadotropina/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Perfusão , Fenótipo , Hipófise/fisiologia , Fatores de Tempo
7.
Comp Biochem Physiol B Biochem Mol Biol ; 129(2-3): 517-24, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11399487

RESUMO

The role of mitogen-activated protein kinase (MAPK, also known as extracellular signal regulated kinase; ERK) stimulation in gonadotropin-releasing hormone (GnRH) signaling was investigated in cultured pituitary cells of tilapia hybrids (Oreochromis niloticus x O. aureus). Exposure of the cells to salmon GnRH (sGnRH) resulted in a dose- and time-dependent elevation in ERK levels. The PKC activator, 1-O-tetradecanoyl phorbol-13-acetate (TPA) increased kinase levels, while addition of GnRH had no further effect. However, chronic exposure to TPA resulted in reduction of basal and GnRH-induced ERK elevation. When PKC was inhibited by GF109203X, the GnRH-elevated ERK levels were totally abolished. The role of MAPK activation on GPalpha, FSHbeta and LHbeta gene expression was determined by administration of MAPK-kinase (MEK) inhibitor (PD98059; PD). This inhibitor completely blocked GnRH-induced increases in ERK activity. Furthermore, it suppressed GPalpha and LHbeta mRNA responses to GnRH, but had no effect on FSHbeta transcript levels. PD also decreased basal LHbeta mRNA levels. These results indicate that in tilapia pituitary cells, GnRH activates MAPK cascade in a PKC-dependent manner. ERK is involved in GnRH elevation of GPalpha and LHbeta, but not in FSHbeta genes transcription.


Assuntos
Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores LHRH/metabolismo , Tilápia/metabolismo , Animais , Western Blotting , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Hormônio Foliculoestimulante/genética , Subunidade beta do Hormônio Folículoestimulante , Regulação da Expressão Gênica/efeitos dos fármacos , Subunidade alfa de Hormônios Glicoproteicos/genética , Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas/genética , Hibridização Genética , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Hormônio Luteinizante/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Subunidades Proteicas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Salmão , Fatores de Tempo
8.
Vitam Horm ; 63: 63-90, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11358118

RESUMO

Gonadotropin releasing hormone (GnRH), the first key hormone of reproduction, is synthesized and secreted from the hypothalamus in a pulsatile manner and stimulates pituitary gonadotrophs (5-10% of the pituitary cells) to synthesize and release gonadotropin luteinizing hormone (LH) and follicle stimulating hormone (FSH). Gonadotrophs consist of 60% multihormonal cells (LH+FSH) and 18% LH- and 22% FSH-containing cells. LH and FSH, members of the glycoprotein hormone family, stimulate spermatogenesis, folliculogenesis, and ovulation. Although GnRH plays a pivotal role in gonadotropin synthesis and release, other factors such as gonadal steroids and gonadal peptides exert positive and negative feedback mechanisms, which affect GnRH actions. GnRH actions include activation of phosphoinositide turnover as well as phospholipase D and A2, mobilization and influx of Ca2+, activation of protein kinase C (PKC) and mitogen-activated protein kinase (MAPK). A complex crosstalk between the above messenger molecules mediates the diverse actions of GnRH. Understanding the signaling mechanisms involved in GnRH actions is the basis for our understanding of basic reproductive functions in general and gonadotropin synthesis and release in particular.


Assuntos
Gonadotropinas/fisiologia , Receptores LHRH/fisiologia , Animais , Ácido Araquidônico/fisiologia , Cálcio/fisiologia , Expressão Gênica , Gonadotropinas/genética , Gonadotropinas/metabolismo , Humanos , Fosfatidilinositóis/metabolismo , Fosfolipases/metabolismo , Hipófise/fisiologia , Proteína Quinase C/fisiologia , Receptores LHRH/química , Transdução de Sinais/fisiologia , Relação Estrutura-Atividade
9.
Cell Mol Neurobiol ; 21(6): 657-74, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12043840

RESUMO

Endothelins have been implicated in the regulation of cell proliferation, differentiation, and apoptosis, but the mechanisms of these complex events are not yet fully understood. Although the nuclear factor-kappaB (NF-kappaB) was shown to play a prominent role in the above processes, its participation in endothelin receptor A (ET(A)R) signaling has not been previously demonstrated. This study provides evidence that NF-kappaB is involved in ET(A)R-induced proliferation and inhibition of apoptosis. Endothelin (ET)-1, ET-3, and sarafotoxin b induce cell proliferation and prevent apoptosis induced by serum deprivation in a Chinese hamster lung (CCL39) cell line that stably expresses ET(A)R (CCL39ET(A)). Activation of ET(A)R resulted in enhanced DNA-binding activity of NF-kappaB and degradation of IkappaB-alpha. Expression of the dominant negative form of IkappaB-alpha (IkappaB deltaN) inhibited the proliferative activities mediated by ET(A)R as well as its anti-apoptotic activities. Treatment of the cells with prostaglandin A1, an inhibitor of IkappaB kinase-beta, reduced ET-1-induced proliferation and its anti-apoptotic effect. These findings indicate that the regulation of cell proliferation and apoptosis by ET(A)R is mediated by the ET(A)R-activated NF-kappaB.


Assuntos
Apoptose/fisiologia , Divisão Celular/fisiologia , Endotelinas/metabolismo , Fibroblastos/metabolismo , Proteínas I-kappa B , NF-kappa B/metabolismo , Receptores de Endotelina/metabolismo , Venenos de Víboras/metabolismo , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Meios de Cultura Livres de Soro/farmacologia , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Endotelina-1/metabolismo , Endotelina-1/farmacologia , Endotelinas/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Mutação/genética , Inibidor de NF-kappaB alfa , Prostaglandinas A/farmacologia , Receptor de Endotelina A , Receptores de Endotelina/agonistas , Venenos de Víboras/farmacologia
10.
J Biol Chem ; 276(7): 4554-63, 2001 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-11083862

RESUMO

G-protein-coupled receptors are a large group of integral membranal receptors, which in response to ligand binding initiate diverse downstream signaling. Here we studied the gonadotropin-releasing hormone (GnRH) receptor, which uses Gq for its downstream signaling. We show that extracellular signal-regulated kinase (ERK) activation is fully dependent on protein kinase C (PKC), but only partially dependent on Src, dynamin, and Ras. Receptor tyrosine kinases, FAK, Gbetagamma, and beta-arrestin, which were implicated in some G-protein-coupled receptor signaling to MAPK cascades, do not play a role in the GnRH to ERK pathway. Our results suggest that the activation of ERK by GnRH involves two distinct signaling pathways, which converge at the level of Raf-1. The main pathway involves a direct activation of Raf-1 by PKC, and this step is partially dependent on a second pathway consisting of Ras activation, which occurs in a dynamin-dependent manner, downstream of Src.


Assuntos
GTP Fosfo-Hidrolases/fisiologia , Hormônio Liberador de Gonadotropina/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Animais , Arrestinas/fisiologia , Linhagem Celular , Dinaminas , Receptores ErbB/fisiologia , Proteína-Tirosina Quinases de Adesão Focal , Proteínas Heterotriméricas de Ligação ao GTP/fisiologia , Sistema de Sinalização das MAP Quinases , Modelos Biológicos , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-raf/metabolismo , beta-Arrestinas
11.
Trends Endocrinol Metab ; 11(3): 91-9, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10707049

RESUMO

G-protein-coupled receptors (GPCRs) are a large group of integral membrane receptors that transmit signals from a diverse array of external stimuli, including neurotransmitters, hormones, phospholipids, photons, odorants and taste ligands. In response to ligand binding, the GPCRs initiate diverse downstream signaling pathways through four groups of G proteins and other interacting proteins. Key components in GPCR-induced intracellular signaling are four groups of mitogen-activated protein kinase (MAPK) cascades: extracellular signal-related kinase (ERK), Jun N-terminal kinase (JNK), p38MAPK and big MAPK (BMK). The hallmark of MAPK signaling is the stimulation-dependent nuclear translocation of the involved kinases, which regulate gene expression and the cytoplasmic acute response to mitogenic, stress-related, apoptotic and survival stimuli. A special type of GPCR is the gonadotropin-releasing hormone (GnRH) receptor, which uses primarily the Gq protein for its downstream signaling. GnRH activates all four MAPK cascades by a PKC-dependent mechanism. Common signaling molecules, including the tyrosine kinase c-SRC and the small GTPases CDC42, RAC and RAS, are implicated in various aspects of the GnRH-MAPK pathways. Thus, the activation of MAPK cascades by GnRH opens a new vista in the understanding of the transcriptional regulation of genes encoding gonadotropins. However, additional studies on cell lines and whole animals are required to understand GnRH signaling in the context of other hormones during the reproductive cycle of mouse and human.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores LHRH/fisiologia , Animais , Ativação Enzimática/fisiologia , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Receptores Proteína Tirosina Quinases/fisiologia , Receptores de Superfície Celular/fisiologia
12.
Rev Reprod ; 4(3): 151-9, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10521152

RESUMO

Binding to the zona pellucida of an egg stimulates the spermatozoon to undergo the acrosome reaction, a process that enables it to penetrate the egg. Before this binding, the spermatozoon undergoes a series of biochemical transformations in the female reproductive tract, collectively called capacitation. Only capacitated spermatozoa can bind to the zona pellucida and undergo the acrosome reaction. Protein kinases may be involved in the regulation of intracellular Ca2+ during capacitation and the acrosome reaction. The first event in capacitation is the increase in intracellular calcium, bicarbonate and hydrogen peroxide, which collectively activate adenylyl cyclase to produce cyclic AMP, which activates protein kinase A to phosphorylate certain proteins. During capacitation, there is an increase in membrane-bound phospholipase C, and this binding is highly stimulated by the addition of epidermal growth factor to the cells. The capacitated spermatozoon binds to the zona pellucida of the egg via specific receptors and it is suggested that the zona pellucida binds to at least two different receptors in the sperm head plasma membrane. One is a Gi-coupled receptor that can activate phospholipase Cbeta1 and may regulate adenylyl cyclase to further increase cyclic AMP concentrations. The cyclic AMP activates protein kinase A to open a calcium channel in the outer acrosomal membrane, resulting in a relatively small increase in cytosolic calcium. This increase in Ca2+ leads to activation of phospholipase Cgamma, which is coupled to the second tyrosine kinase receptor. The products of phosphatidyl-inositol bisphosphate hydrolysis by phospholipase C, diacylglycerol and inositol-trisphosphate, induce the activation of protein kinase C and a calcium channel in the outer acrosomal membrane, respectively. Protein kinase C opens a calcium channel in the plasma membrane and, together with the inositol-trisphosphate-activated calcium channel, leads to a second and higher increase in cytosolic calcium. In addition, the depletion of calcium in the acrosome activates a capacitative calcium entry mechanism in the plasma membrane, leading to a rapid increase in cytosolic calcium (300-500 nmol l(-1)). This increase in intracellular calcium concentration (and pH) leads to membrane fusion and the acrosome reaction.


Assuntos
Reação Acrossômica , Mamíferos/metabolismo , Proteínas Quinases/metabolismo , Capacitação Espermática , Animais , ATPase de Ca(2+) e Mg(2+) , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Humanos , Masculino , Proteínas Tirosina Quinases/metabolismo , Interações Espermatozoide-Óvulo
13.
Chronobiol Int ; 16(4): 441-50, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10442238

RESUMO

In the present study, we examined in vitro luteinizing hormone (LH) release patterns from pituitaries and from pituitary cell cultures (3 and 7 days in culture) to elucidate the endogenous period generated by the gonadotroph cell population and to evaluate the relationship between the basic period generated at the cellular level and the output pattern observed at the organ level. In addition, we examined the effect of photic environmental signals perceived by the animals on LH release patterns from pituitaries in vitro. When the animals were exposed to circadian photoperiodic signals, the in vitro LH release pattern from the pituitaries exhibited ultradian, circadian, and infradian frequencies. When the animals were exposed to continuous illumination, the in vitro patterns exhibited only ultradian and infradian frequencies. Furthermore, free running is a process, not a state. This process is driven by a change in the relative dominance of different frequencies that construct the pattern without changing the basic period length. Evaluation of the relative dominance of the different frequencies that construct the pattern indicates that, although infradian oscillators may take part in shaping the output pattern, the basic rhythm generated by the pituitary cells is in the ultradian domain. The results obtained from the examined system suggest that an endogenous oscillator is a cellular entity with ultradian periodicity, and that the rhythmic output of many biological variables is structured by various ultradian components that construct the circadian and infradian output rhythms.


Assuntos
Ciclos de Atividade/fisiologia , Hormônio Luteinizante/metabolismo , Periodicidade , Animais , Células Cultivadas , Ritmo Circadiano/fisiologia , Feminino , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos ICR , Fotoperíodo , Hipófise/metabolismo
14.
Ann Endocrinol (Paris) ; 60(2): 79-88, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10456177

RESUMO

The purpose of this review is to update the information concerning the intracellular effect of GnRH. Binding of GnRH to a G-protein coupled receptor leads to stimulation of Gq and/or G11 protein and to activation of phospholipase C beta. Inositol 1-4-5-triphosphate and early diacylylycerol are the second messengers required for conventional protein kinase C activation. Activation of phospholipase A2 and phospholipase D are also involved, as demonstrated by the liberation of Arachidonic Acid and Phosphatidic Acid. Pituitary cells also express atypical protein kinase C isoforms which mode of activation is not known. Hypothesis concerning transcriptional regulation are presented.


Assuntos
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Receptores LHRH/fisiologia , Transdução de Sinais , Animais , Proteínas de Ligação ao GTP/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Fosfolipases/metabolismo , Proteínas Quinases/metabolismo , Receptores LHRH/química
15.
J Biol Chem ; 274(11): 7431-40, 1999 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-10066808

RESUMO

Evidence for depolarization-induced activation of G-proteins in membranes of rat brain synaptoneurosomes has been previously reported (Cohen-Armon, M., and Sokolovsky, M. (1991) J. Biol. Chem. 266, 2595-2605; Cohen-Armon, M., and Sokolovsky, M. (1993) J. Biol. Chem. 268, 9824-9838). In the present work we identify the activated G-proteins as Go-proteins by tracing their depolarization-induced in situ photoaffinity labeling with [alpha32P]GTP-azidoanilide (GTPAA). Labeled GTPAA was introduced into transiently permeabilized rat brain-stem synaptoneurosomes. The resealed synaptoneurosomes, while being UV-irradiated, were depolarized. Relative to synaptoneurosomes at resting potential, the covalent binding of [alpha32P]GTPAA to Galphao1- and Galphao3-proteins, but not to Galphao2- isoforms, was enhanced by 5- to 7-fold in depolarized synaptoneurosomes, thereby implying an accelerated exchange of GDP for [alpha32P]GTPAA. Their depolarization-induced photoaffinity labeling was independent of stimulation of Go-protein-coupled receptors and could be reversed by membrane repolarization, thus excluding induction by transmitters release. It was, however, dependent on depolarization-induced activation of the voltage-gated sodium channels (VGSC), regardless of Na+ current. The alpha subunit of VGSC was cross-linked and co-immunoprecipitated with Galphao-proteins in depolarized brain-stem and cortical synaptoneurosomes. VGSC alpha subunit most efficiently cross-linked with guanosine 5'-O-2-thiodiphosphate-bound rather than to guanosine 5'-O-(3-thiotriphosphate)-bound Galphao-proteins in isolated synaptoneurosomal membranes. These findings support a possible involvement of VGSC in depolarization-induced activation of Go-proteins.


Assuntos
Azidas/química , Proteínas de Ligação ao GTP/metabolismo , Guanosina Trifosfato/análogos & derivados , Potenciais da Membrana , Marcadores de Fotoafinidade/química , Adenosina Difosfato Ribose/metabolismo , Animais , Proteínas de Ligação ao GTP/química , Guanosina Trifosfato/química , Ativação do Canal Iônico , Masculino , Fosforilação , Testes de Precipitina , Ligação Proteica , Ratos , Receptores de Neurotransmissores/metabolismo , Canais de Sódio/metabolismo
16.
Mol Endocrinol ; 12(6): 815-24, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9626657

RESUMO

The signaling of ligands operating via heterotrimeric G proteins is mediated by a complex network that involves sequential phosphorylation events. Signaling by the G protein-coupled receptor GnRH was shown to include elevation of Ca2+ and activation of phospholipases, protein kinase C (PKC) and extra-cellular signal-regulated kinase (ERK). In this study, GnRH was shown to activate Jun N-Terminal Kinase (JNK)/SAPK in alpha T3-1 cells in a PKC- and tyrosine kinase-dependent manner. GnRH as well as tumor-promoting agent (TPA) also increased c-Src activity, which peaked at 2 min after GnRH stimulation and was sensitive both to PKC and to tyrosine kinase inhibitors. Coexpression of Csk, which serves as a Src-dominant interfering kinase, and constitutively active forms of Src, together with JNK, confirmed the involvement of c-Src downstream of PKC in the GnRH-JNK pathway. Coexpression of dominant negative and constitutively active forms of CDC42, Rac1, Ras, MEKK1, and MEK1 with JNK indicated that JNK activation by GnRH and TPA is mediated by CDC42 and MEKK1. Ras and MEK1, which are involved in a related mitogen-activated protein kinase (MAPK) pathway, did not affect JNK activation in alpha T3-1 cells. Taken together, our results suggest that GnRH stimulation of JNK activity is mediated by a unique pathway that includes sequential activation of PKC, c-Src, CDC42, and probably also MEKK1.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas de Ciclo Celular/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Hormônio Liberador de Gonadotropina/farmacologia , MAP Quinase Quinase Quinase 1 , Proteínas Quinases Ativadas por Mitógeno , Adeno-Hipófise/efeitos dos fármacos , Proteína Quinase C/fisiologia , Proteínas Proto-Oncogênicas pp60(c-src)/fisiologia , Transdução de Sinais/fisiologia , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Proteína Quinase 1 Ativada por Mitógeno , Proteína Quinase 3 Ativada por Mitógeno , Adeno-Hipófise/citologia , Adeno-Hipófise/enzimologia , Proteína Quinase C/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Recombinantes de Fusão/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica , Transfecção , Proteína cdc42 de Ligação ao GTP , Proteínas rac de Ligação ao GTP
17.
Am J Physiol ; 274(2): E218-23, 1998 02.
Artigo em Inglês | MEDLINE | ID: mdl-9486150

RESUMO

Acrosomal exocytosis in mammalian spermatozoa is a process essential for fertilization. We report here that atrial natriuretic peptide (ANP) markedly stimulates acrosomal exocytosis of capacitated human spermatozoa. Typically, ANP exerts some of its actions via activation of the ANP receptor (ANPR-A), a particulate guanylyl cyclase-linked receptor, and subsequent formation of guanosine 3',5'-cyclic monophosphate (cGMP). We found that ANP-stimulated acrosome reaction was inhibited by the competitive ANPR-A antagonist anantin, indicating a receptor-mediated process. A linear fragment of ANP, ANP-(13-28), and another ANP-like compound, brain natriuretic peptide, were inactive. The stimulatory effect of ANP on acrosome reaction was mimicked by the permeable cGMP analog, 8-bromo-cGMP (8-BrcGMP). Addition of the protein kinase C (PKC) inhibitors, staurosporine and GF-109203X, resulted in a dose-related inhibition of ANP-induced acrosome reaction. Also, downregulation of endogeneous PKC activity resulted in inhibition of ANP- but not 8-BrcGMP-induced acrosome reaction. Removal of extracellular Ca2+ abolished ANP-induced acrosome reaction. Thus ANP via Ca2+ influx, PKC activation, and stimulation of particulate guanylyl cyclase may play a role in the induction of acrosome reaction of human spermatozoa.


Assuntos
Fator Natriurético Atrial/fisiologia , Exocitose/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Acrossomo/efeitos dos fármacos , Adulto , Animais , Cálcio/metabolismo , GMP Cíclico/metabolismo , Humanos , Masculino , Proteína Quinase C/metabolismo , Ratos , Receptores do Fator Natriurético Atrial/metabolismo , Capacitação Espermática/efeitos dos fármacos , Espermatozoides/fisiologia
18.
Front Neuroendocrinol ; 19(1): 1-19, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9465287

RESUMO

Gonadotropin-releasing hormone (GnRH), the first key hormone of reproduction, is synthesized in the hypothalamus and is released in a pulsatile manner to stimulate pituitary gonadotrope-luteinizing hormone (LH) and follicle-stimulating hormone (FSH) synthesis and release. Gonadotropes represent only about 10% of pituitary cells and are divided into monohormonal cells (18% LH and 22% FSH cells) and 60% multihormonal (LH + FSH) cells. GnRH binds to a specific seven transmembrane domain receptor which is coupled to Gq and activates sequentially different phospholipases to provide Ca2+ and lipid-derived messenger molecules. Initially, phospholipase C is activated, followed by activation of both phospholipase A2 (PLA2) and phospholipase D (PLD). Generation of the second messengers inositol 1,4,5-trisphosphate and diacylglycerol (DAG) lead to mobilization of intracellular pools of Ca2+ and activation of protein kinase C (PKC). Early DAG and Ca2+, derived via enhanced phosphoinositide turnover, might be involved in rapid activation of selective Ca(2+)-dependent, conventional PKC isoforms (cPKC). On the other hand, late DAG, derived from phosphatidic acid (PA) via PLD, may activate Ca(2+)-independent novel PKC isoforms (nPKC). In addition, arachidonic acid (AA) which is liberated by activated PLA2, might also support selective activation of PKC isoforms (PKCs) with or without other cofactors. Differential cross-talk of Ca2+, AA, and selective PKCs might generate a compartmentalized signal transduction cascade to downstream elements which are activated during the neurohormone action. Among those elements is the mitogen-activated protein kinase (MAPK) cascade which is activated by GnRH in a PKC-, Ca(2+)-, and protein tyrosine kinase (PTK)-dependent fashion. Transcriptional regulation can be mediated by the activation of transcription factors such as c-fos by MAPK. Indeed, GnRH activates the expression of both c-jun and c-fos which might participate in gene regulation via the formation of AP-1. The signaling cascade leading to gonadotropin (LH and FSH) gene regulation by GnRH is still not known and might involve the above-mentioned cascades. AA and selective lipoxygenase products such as leukotriene C4 also participate in GnRH action, possibly by cross-talk with PKCs, or by an autocrine/paracrine amplification cycle. A complex combinatorial, spatial and temporal cross-talk of the above messenger molecules seems to mediate the diverse effects elicited by GnRH, the first key hormone of the reproductive cycle.


Assuntos
Cálcio/fisiologia , Proteína Quinase C/fisiologia , Receptores LHRH/fisiologia , Transdução de Sinais/fisiologia , Animais , Hormônio Liberador de Gonadotropina/fisiologia , Humanos , Receptores LHRH/química , Relação Estrutura-Atividade
19.
Trends Endocrinol Metab ; 9(7): 297, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18406289
20.
J Biol Chem ; 272(21): 13534-40, 1997 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-9153199

RESUMO

The effect of gonadotropin-releasing hormone (GnRH) upon protein kinase C (PKC) delta and PKCepsilon gene expression was investigated in the gonadotroph-derived alphaT3-1 cell line. Stimulation of the cells with a stable analog [D-Trp6]GnRH (GnRH-A) resulted in a rapid elevation of PKCepsilon mRNA levels (1 h), while PKCdelta mRNA levels were elevated only after 24 h of incubation. The rapid elevation of PKCepsilon mRNA by GnRH-A was blocked by pretreatment with a GnRH antagonist or actinomycin D. The PKC activator 12-O-tetradecanoylphorbol-13-acetate (TPA), but not the Ca2+ ionophore ionomycin, mimicked the rapid effect of GnRH-A upon PKCepsilon mRNA elevation. Additionally, the rapid stimulatory effect of GnRH-A was blocked by the selective PKC inhibitor GF109203X, by TPA-mediated down-regulation of endogenous PKC, or by Ca2+ removal. Interestingly, serum-starvation (24 h) advanced the stimulation of PKCdelta mRNA levels by GnRH-A and the effect could be detected at 1 h of incubation. The rapid effect of GnRH-A upon PKCdelta mRNA levels in serum-starved cells was mimicked by TPA, but not by ionomycin, and was abolished by down-regulation of PKC or by Ca2+ removal. Preactivation of alphaT3-1 cells with GnRH-A for 1 h followed by removal of ligand and serum resulted in elevation of PKCdelta mRNA levels after 24 h of incubation. Western blot analysis revealed that GnRH-A and TPA stimulated (within 5 min) the activation and some degradation of PKCdelta and PKCepsilon. We conclude that Ca2+ and PKC are involved in GnRH-A elevation of PKCdelta and PKCepsilon mRNA levels, with Ca2+ being necessary but not sufficient, while PKC is both necessary and sufficient to mediate the GnRH-A response. A serum factor masks PKCdelta but not PKCepsilon mRNA elevation by GnRH-A, and its removal exposes preactivation of PKCdelta mRNA by GnRH-A which can be memorized for 24 h. PKCdelta and PKCepsilon gene expression evoked by GnRH-A is autoregulated by PKC, and both isotypes might participate in the neurohormone action.


Assuntos
Regulação Enzimológica da Expressão Gênica , Hormônio Liberador de Gonadotropina/fisiologia , Homeostase , Isoenzimas/genética , Proteína Quinase C/genética , Animais , Cálcio/metabolismo , Carcinógenos/farmacologia , Linhagem Celular , Ativação Enzimática , Ionomicina/farmacologia , Ionóforos/metabolismo , Isoenzimas/fisiologia , Hipófise/citologia , Hipófise/enzimologia , Proteína Quinase C/fisiologia , Proteína Quinase C-delta , Proteína Quinase C-épsilon , RNA Mensageiro/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...