Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
2.
Mol Endocrinol ; 1(11): 808-15, 1987 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-3155261

RESUMO

GnRH releases LH from pituitary gonadotropes by a calcium-dependent mechanism. Previous studies in static cell cultures have not revealed a role for intracellular-derived calcium during GnRH-stimulated LH release. In the present study we have reexamined this possibility using a perifusion system, which permits a more dynamic assessment of early cellular events. Chelation of extracellular calcium by EGTA and calcium channel blockade by methoxyverapamil prevented sustained LH release. A component of early LH release occurred independently of extracellular calcium mobilization. This previously unrecognized aspect of LH release was shown to be dependent upon intracellular calcium. The molecular mechanism by which this calcium-dependent signal is translated into a cellular response does not appear to be mediated by calmodulin or protein kinase C, whereas sustained LH release appears mediated by calmodulin. While calcium derived from extracellular sources is still viewed as the major messenger for sustained LH release, these experiments provide evidence for the involvement of intracellular-derived calcium during early GnRH-stimulated LH release.


Assuntos
Cálcio/fisiologia , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Animais , Calcimicina/farmacologia , Células Cultivadas , Ácido Egtázico/farmacologia , Feminino , Galopamil/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante/análise , Penfluridol/farmacologia , Perfusão , Hipófise/citologia , Proteína Quinase C/metabolismo , Radioimunoensaio , Ratos , Ratos Endogâmicos , Acetato de Tetradecanoilforbol/farmacologia
3.
Mol Endocrinol ; 11(6): 738-46, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9171237

RESUMO

Evidence from use of pertussis and cholera toxins and from NaF suggested the involvement of G proteins in GnRH regulation of gonadotrope function. We have used three different methods to assess GnRH receptor regulation of G(q/11)alpha subunits (G(q/11)alpha). First, we used GnRH-stimulated palmitoylation of G(q/11)alpha to identify their involvement in GnRH receptor-mediated signal transduction. Dispersed rat pituitary cell cultures were labeled with [9,10-(3)H(N)]-palmitic acid and immunoprecipitated with rabbit polyclonal antiserum made against the C-terminal sequence of G(q/11)alpha. The immunoprecipitates were resolved by 10% SDS-PAGE and quantified. Treatment with GnRH resulted in time-dependent (0-120 min) labeling of G(q/11)alpha. GnRH (10(-12), 10(-10), 10(-8), or 10(-6) g/ml) for 40 min resulted in dose-dependent labeling of G(q/11)alpha compared with controls. Cholera toxin (5 microg/ml; activator of G(i)alpha), pertussis toxin (100 ng/ml; inhibitor of G(i)alpha actions) and Antide (50 nM; GnRH antagonist) did not stimulate palmitoylation of G(q/11)alpha above basal levels. However, phorbol myristic acid (100 ng/ml; protein kinase C activator) stimulated the palmitoylation of G(q/11)alpha above basal levels, but not to the same extent as 10(-6) g/ml GnRH. Second, we used the ability of the third intracellular loop (3i) of other seven-transmembrane segment receptors that couple to specific G proteins to antagonize GnRH receptor-stimulated signal transduction and therefore act as an intracellular inhibitor. Because the third intracellular loop of alpha1B-adrenergic receptor (alpha1B 3i) couples to G(q/11)alpha, it can inhibit G(q/11)alpha-mediated stimulation of inositol phosphate (IP) turnover by interfering with receptor coupling to G(q/11)alpha. Transfection (efficiency 5-7%) with alpha1B 3i cDNA, but not the third intracellular loop of M1-acetylcholine receptor (which also couples to G(q/11)alpha), resulted in 10-12% inhibition of maximal GnRH-evoked IP turnover, as compared with vector-transfected GnRH-stimulated IP turnover. The third intracellular loop of alpha2A adrenergic receptor, M2-acetylcholine receptor (both couple to G(i)alpha), and D1A-receptor (couples to G(s)alpha) did not inhibit IP turnover significantly compared with control values. GnRH-stimulated LH release was not affected by the expression of these peptides. Third, we assessed GnRH receptor regulation of G(q/11)alpha in a PRL-secreting adenoma cell line (GGH(3)1') expressing the GnRH receptor. Stimulation of GGH(3)1' cells with 0.1 microg/ml Buserelin (a metabolically stable GnRH agonist) resulted in a 15-20% decrease in total G(q/11)alpha at 24 h following agonist treatment compared with control levels; this action of the agonist was blocked by GnRH antagonist, Antide (10(-6) g/ml). Neither Antide (10(-6) g/ml, 24 h) alone nor phorbol myristic acid (0.33-100 ng/ml, 24 h) mimicked the action of GnRH agonist on the loss of G(q/11)alpha immunoreactivity. The loss of G(q/11)alpha immunoreactivity was not due to an effect of Buserelin on cell-doubling times. These studies provide the first direct evidence for regulation of G(q/11)alpha by the GnRH receptor in primary pituitary cultures and in GGH3 cells.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Receptores LHRH/metabolismo , Animais , Busserrelina/farmacologia , Células Cultivadas , Toxina da Cólera/farmacologia , Feminino , Oligopeptídeos/farmacologia , Palmitatos/farmacocinética , Toxina Pertussis , Hipófise/citologia , Hipófise/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Receptores de Dopamina D1/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Virulência de Bordetella/farmacologia
4.
Mol Endocrinol ; 1(2): 154-9, 1987 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2457798

RESUMO

Maitotoxin (MTX) stimulates gonadotropin release from pituitary cell cultures. The time course and efficacy of LH release in response to GnRH and to MTX are similar; both secretagogues require extracellular Ca2+ and are inhibited by the selective Ca2+ ion channel antagonist methoxyverapamil (D600). LH release in response to either GnRH or MTX is not measurably inhibited by two other chemical classes of Ca2+ ion channel inhibitors represented by nifedipine and by diltiazem. The two secretagogues are nonadditive in their action on LH release when presented at high doses and prior studies indicate that MTX has no endogenous ionophoretic activity. These observations indicate that MTX likely stimulates LH release due to activation of the GnRH receptor associated Ca2+-ion channel in the gonadotrope. We have therefore assessed the functional state of this channel during the development of homologous desensitization of the gonadotrope to GnRH by measuring the ability of MTX to stimulate LH release. Cells were desensitized with GnRH in the presence of 3 mM EGTA. Under these conditions, the cells become refractory to GnRH in the absence of gonadotropin release since the latter process, but not the former, requires extracellular Ca2+. Accordingly, this approach allows assessment of the degree of desensitization in the absence of the influence of gonadotropin depletion. Such desensitized cells are less responsive to GnRH. Desensitized pituitary cells also respond with diminished efficacy and potency to MTX three or more hours after GnRH treatment but not at an earlier time (1 h) when GnRH receptors are diminished. These data are consistent with a model in which homologous desensitization is viewed as developing in two phases.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Gonadotropinas Hipofisárias/imunologia , Canais Iônicos/fisiologia , Toxinas Marinhas/farmacologia , Oxocinas , Adeno-Hipófise/efeitos dos fármacos , Hormônios Liberadores de Hormônios Hipofisários/farmacologia , Receptores LHRH/efeitos dos fármacos , Animais , Células Cultivadas , Dessensibilização Imunológica , Feminino , Canais Iônicos/metabolismo , Adeno-Hipófise/citologia , Ratos , Ratos Endogâmicos
5.
Mol Endocrinol ; 1(6): 420-9, 1987 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2856305

RESUMO

The demonstration that GnRH provokes the accumulation of diacylglycerol and the redistribution of protein kinase C to the membrane fraction in gonadotropes suggests a role for this enzyme as a mediator of GnRH action. In the present work we have investigated the possibility that protein kinase C might mediate GnRH-stimulated receptor down-regulation and desensitization. Pretreatment of pituitary cells for 6 h with GnRH (10(-11) - 10(-6) M) caused a biphasic change in GnRH receptor number [the maximum binding (Bmax) for 125I-buserelin binding was increased by 10(-10) M GnRH and reduced by 10(-7) and 10(-6) M GnRH] and caused desensitization (pretreatment with 10(-9) - 10(-6) M GnRH reduced the proportion of cellular LH released in a subsequent challenge with GnRH). Pretreatment for 6 h with 0.2-200 nM phorbol myristate acetate (a protein kinase C-activating phorbol ester) did not cause desensitization, but at 200 nM, did reduce GnRH receptor number. As a further test of the requirement for protein kinase C for GnRH action, cells were depleted of all measurable protein kinase C (and rendered unresponsive to protein kinase C activators) by prior treatment with a high dose of phorbol myristate acetate (500 nM for 6 h followed by 12 h in plating medium). Depletion of protein kinase C did not alter the ability of GnRH to desensitize gonadotropes or down-regulate its own receptors. The demonstration that the effects of GnRH on receptor number and gonadotrope responsiveness are neither blocked by depletion of protein kinase C nor entirely mimicked by activation of protein kinase C suggests that these effects of the releasing hormone are not solely mediated by this enzyme.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/farmacologia , Hipófise/metabolismo , Proteína Quinase C/metabolismo , Receptores LHRH/metabolismo , Animais , Busserrelina/metabolismo , Feminino , Cinética , Ratos , Ratos Endogâmicos , Receptores LHRH/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia
6.
Mol Endocrinol ; 8(8): 1038-48, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7527898

RESUMO

TRH and GnRH receptors are each coupled to G proteins of the Gq/11 family. Activation of each of these receptors by their respective ligands results in the stimulation of phospholipase C activity, leading to calcium mobilization and protein kinase C activation. Thus, the effects of TRH and GnRH may be mediated through the same intracellular signal transduction pathway. To compare responses to TRH and GnRH directly within one cell type, we have stably transfected the rat pituitary GH3 lactotrope cell line, which expresses the endogenous TRH receptor, with an expression vector containing rat GnRH receptor cDNA. Transfected cells specifically bound GnRH with high affinity and responded to GnRH stimulation with an increase in PRL mRNA levels, analogous to their response to TRH stimulation. Stably transfected GH3 cells, which were then transiently transfected with luciferase reporter constructs containing either the PRL or the glycoprotein hormone alpha-subunit promoter, responded to either GnRH or TRH stimulation with an increase in luciferase activity in a time- and dose-dependent fashion. The stimulatory effects of maximally effective concentrations of TRH and GnRH were additive on PRL, but not alpha-subunit, gene expression. These data, coupled with evidence of cross-desensitization of alpha-subunit, but not PRL, promoter activity stimulation by TRH and GnRH, suggest that there may be differences in the signal transduction pathways activated by TRH and GnRH receptors in the regulation of PRL and alpha-subunit gene expression.


Assuntos
Adenoma/patologia , Proteínas de Ligação ao GTP/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/farmacologia , Neoplasias Hipofisárias/patologia , Receptores LHRH/fisiologia , Receptores do Hormônio Liberador da Tireotropina/fisiologia , Transdução de Sinais , Hormônio Liberador de Tireotropina/farmacologia , Animais , Busserrelina/farmacologia , Cálcio/fisiologia , DNA Complementar/genética , Subunidade alfa de Hormônios Glicoproteicos/biossíntese , Subunidade alfa de Hormônios Glicoproteicos/genética , Hormônio Liberador de Gonadotropina/análogos & derivados , Luciferases/biossíntese , Fosfatidilinositol Diacilglicerol-Liase , Diester Fosfórico Hidrolases/fisiologia , Prolactina/genética , Prolactina/metabolismo , Proteína Quinase C/fisiologia , Ratos , Receptores LHRH/efeitos dos fármacos , Receptores do Hormônio Liberador da Tireotropina/efeitos dos fármacos , Proteínas Recombinantes de Fusão/biossíntese , Transfecção , Células Tumorais Cultivadas
7.
Mol Endocrinol ; 12(2): 161-71, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9482659

RESUMO

Mammalian GnRH receptor (GnRHR) is unique among G protein-coupled seven-transmembrane segment receptors due to the absence of an intracellular C-terminal tail frequently important for internalization and/or desensitization of other G protein-coupled receptors. The recent cloning of nonmammalian (i.e. catfish, goldfish, frog, and chicken) GnRHRs shows that these contain an intracellular C terminus. Addition of the 51-amino acid intracellular C terminus from catfish GnRHR (cfGnRHR) to rat GnRHR (rGnRHR) did not affect rGnRHR binding affinity but elevated receptor expression by about 5-fold. Truncation of the added C terminus impaired the elevated receptor-binding sites by 3- to 8-fold, depending on the truncation site. In addition, introducing the C terminus to rGnRHR altered the pattern of receptor regulation from biphasic down-regulation and recovery to monophasic down-regulation. The extent of down-regulation was also enhanced. The alteration in receptor regulation due to the addition of a C terminus was reversed by truncation of the added C terminus. Furthermore, addition of the cfGnRHR C terminus to rGnRHR significantly augmented the inositol phospholipid (IP) response of transfected cells to Buserelin, but this did not result from the elevation of receptor-binding sites. Addition of the C terminus did not affect Buserelin-stimulated cAMP and PRL release. GH3 cells transfected with wild-type cfGnRHR did not show measurable Buserelin binding or significant stimulation of IP, cAMP, or PRL in response to Buserelin (10[-13]-10[-9] M). GH3 cells transfected with C terminus-truncated cfGnRHR showed no IP response to Buserelin (10[-13]-10[-7] M). These results suggest that addition of the cfGnRHR intracellular C terminus to rGnRHR has a significant impact on rGnRHR expression and regulation and efficiency of differential receptor coupling to G proteins.


Assuntos
Regulação da Expressão Gênica , Receptores LHRH/biossíntese , Receptores LHRH/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/síntese química , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Busserrelina/farmacologia , Peixes-Gato , AMP Cíclico/biossíntese , Regulação para Baixo/genética , Fosfatos de Inositol/biossíntese , Líquido Intracelular/química , Líquido Intracelular/metabolismo , Dados de Sequência Molecular , Prolactina/metabolismo , Ligação Proteica/genética , Ratos , Receptores LHRH/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Homologia de Sequência de Aminoácidos
8.
Endocrinology ; 133(6): 2756-60, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8243300

RESUMO

The present study assesses the relationship between G-proteins and protein kinase-C (PKC) in the gonadotrope. Cells were depleted of PKC with 1 microM phorbol 12-myristate 13-acetate for 12 h, followed by medium 199-BSA for 6 h before treatment with vehicle, pertussis toxin (PTX), cholera toxin (CTX), or (Bu)2cAMP (dBcAMP) for 18 h. PTX (10 ng/ml) significantly decreased GnRH-stimulated inositol phosphate (IP) production over a range of 10(-8)-10(-6) M GnRH. The degree of this inhibition was the same in control cells and PKC-depleted cells. Pretreatment with CTX (0.5 microgram/ml) significantly decreased GnRH-stimulated IP production over a range of 10(-9)-10(-6) M GnRH in PKC-depleted cells. This effect was mimicked by pretreatment with 3 mM dBcAMP. Although CTX and dBcAMP both decreased GnRH-stimulated IP production in control cells, this effect was enhanced in PKC-depleted cells. CTX (0.1 microgram/ml) and dBcAMP (3 mM) both enhanced GnRH-stimulated LH release, whereas PTX (100 ng/ml) had no effect. This was observed in control as well as PKC-depleted cells. Both PKA and PKC are capable of regulating IP turnover by phosphorylating phospholipase-C at distinct sites. CTX activates a G-protein that increases cAMP. cAMP can then activate PKA. In PKC-depleted cells, CTX inhibits GnRH-stimulated IP production. This effect is mimicked by dBcAMP, which suggests a role for PKA in the gonadotrope. The results of this study provide evidence for cross-talk between a CTX-sensitive G-protein and PKC.


Assuntos
Bucladesina/farmacologia , Toxina da Cólera/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Fosfatos de Inositol/biossíntese , Hipófise/metabolismo , Proteína Quinase C/deficiência , Animais , Células Cultivadas , Fosfatos de Inositol/antagonistas & inibidores , Hormônio Luteinizante/metabolismo , Toxina Pertussis , Hipófise/citologia , Ratos , Fatores de Virulência de Bordetella/farmacologia
9.
Endocrinology ; 118(3): 1148-58, 1986 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-3004897

RESUMO

GnRH stimulates rapid and specific hydrolysis of the phosphoinositides and their conversion to diacylglycerols (DAGs) in enriched gonadotrope cultures. In short term labeling studies, we observed stimulation of 32P incorporation into inositol phospholipids; no corresponding change was seen in other major phospholipids. The observation that this response continues in the presence of the Ca2+ channel antagonist D600 suggests that, unlike LH release, stimulation of phosphatidylinositol (PI) production is independent of mobilization of extracellular Ca2+. Agents that substitute for endogenous DAGs (phorbol myristate acetate) or mobilize Ca2+ directly (ionophore A23187) did not stimulate 32P incorporation into PI. Relative mass changes in PIs in response to GnRH were measured in a 32P/33P protocol. These showed a fall (40%) in the level of the inositol phospholipids within 45 sec, with a new steady state achieved at a lower level by 5 min. Phosphatidic acid formation was stimulated by GnRH within 2 min, with a 150% increase above control values at the highest dose of GnRH tested (10(-7) M). In cells prelabeled with [3H]arachidonic acid, GnRH caused a transient increase in DAG formation (twice the control value) within 1 min (with or without D600) which declined thereafter. This effect of GnRH was not seen in cells stimulated to release LH with ionophore A23187 or phorbol myristate acetate, suggesting that these may act distally to inositol phospholipid turnover. The data suggest that GnRH stimulates the hydrolysis of newly synthesized PIs, leading to the formation of DAGs. This pathway appears to be regulated independently of mobilization of extracellular Ca2+.


Assuntos
Diglicerídeos/metabolismo , Glicerídeos/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Fosfatidilinositóis/metabolismo , Adeno-Hipófise/metabolismo , Animais , Ácido Araquidônico , Ácidos Araquidônicos/metabolismo , Células Cultivadas , Feminino , Ácidos Fosfatídicos/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Adeno-Hipófise/efeitos dos fármacos , Ratos
10.
Endocrinology ; 118(6): 2599-604, 1986 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3084223

RESUMO

GnRH stimulates LH release from pituitary gonadotropes. Prolonged exposure of these cells to GnRH results in decreased sensitivity to further stimulation by the releasing hormone both in vivo and in vitro. Chelation of extracellular Ca++ with EGTA blocks GnRH-stimulated LH release but does not prevent subsequent desensitization. Desensitization occurs when cells are preincubated in EGTA containing 10(-7) M GnRH for a variety of times (20 min to 12 h) or when cells are preincubated for 3 h in EGTA with 10(-10), 10(-9), or 10(-8) M GnRH. A GnRH antagonist does not cause desensitization to GnRH and blocks desensitization in response to GnRH in the Ca++-free medium. Preincubation in EGTA containing 10(-7) M GnRH for 3 h did not alter sensitivity of cells to sn 1,2 dioctanoylglycerol (a protein kinase C activator), Ca++ ionophore A23187, or veratridine (an activator of endogenous ion channels). These results suggest that desensitization results from occupancy of the GnRH receptor by an agonist and may be uncoupled from LH release.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante/metabolismo , Adeno-Hipófise/metabolismo , Animais , Calcimicina/farmacologia , Cálcio/fisiologia , Células Cultivadas , Diglicerídeos/farmacologia , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Ácido Egtázico/farmacologia , Feminino , Adeno-Hipófise/efeitos dos fármacos , Ratos , Veratridina/farmacologia
11.
Endocrinology ; 132(5): 2131-5, 1993 May.
Artigo em Inglês | MEDLINE | ID: mdl-8386609

RESUMO

Cholera toxin (CTX; 5 micrograms/ml), but not pertussis toxin (100 ng/ml), when preincubated with pituitary cells for 18 h, enhances the percentage of cellular LH released in response to continuous or pulsatile administration of 5 x 10(-9) M GnRH. This effect occurs without increasing total (intracellular plus extracellular) LH, indicating that it is best explained by redistribution of LH from a nonreleasable to a releasable pool. This site of action is consistent with the observation that CTX-pretreated cells are also sensitized to stimulation of LH release by the Ca2+ ionophore A23187. The observations that CTX stimulates the production of cAMP in these cells and that the sensitizing action of CTX is mimicked by (Bu)2cAMP (1 mM) are consistent with the view that a CTX-stimulated guanyl nucleotide binding protein, capable of activating adenylyl cyclase, is mediating this sensitization. We used a perifused cell system to show that the movement of LH into a releasable pool is lost with the onset of homologous desensitization due to high pulse frequency or constant administration of GnRH (5 x 10(-9) M, continuous or a pulse each 15 min). Sensitization to CTX is restored by stimulation with a high concentration of GnRH (10(-6) M) or by resetting the pulse frequency to the rate measured in vivo (a pulse each 90 min). Both of these treatments also circumvent the desensitized state, restoring LH release. These results identify a novel lesion associated with the development of desensitization in the gonadotrope and support the role of a CTX-sensitive guanyl nucleotide binding protein in regulation of pituitary responsiveness to GnRH.


Assuntos
Toxina da Cólera/farmacologia , Proteínas de Ligação ao GTP/fisiologia , Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Animais , Bucladesina/farmacologia , Calcimicina/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Tolerância a Medicamentos , Feminino , Hormônio Liberador de Gonadotropina/administração & dosagem , Hipófise/efeitos dos fármacos , Ratos
12.
Endocrinology ; 130(4): 2091-100, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1547730

RESUMO

Involvement of ionized cytosolic calcium ([Ca2+]i) and protein kinase-C (PKC) in GnRH-stimulated LH release was assessed by correlating measurable changes in [Ca2+]i and LH release in PKC-depleted and nondepleted gonadotropes. Primary cultures of anterior pituitary cells were loaded with the calcium-sensitive fluorescent dye fura-2 and placed in a perifusion chamber. GnRH pulses were delivered to the cells, and changes in fura-2 fluorescence and LH release were determined. The level of [Ca2+]i (assessed by fura-2) increased rapidly to a maximum within 20-40 sec, followed by a slower decline over the next minute (spike phase) to a sustained intermediate value (plateau phase). GnRH-stimulated LH release was unaffected by loading cells with fura-2. Both LH release and changes in [Ca2+]i were directly dependent on GnRH concentration. Pretreatment with the GnRH antagonist Antide (50 nM; [NAcD2Nal1-DpClPhe2-D3Pal3-Ser4-NicLys5-++ +DNicLys6-Leu7-ILys8-Pro9-DAla10]NH2 ) had no effect on basal [Ca2+]i or basal LH release, but did block both GnRH-stimulated calcium mobilization and GnRH-stimulated LH release. GnRH pretreatment (3.5 nM; 10 min) blocked the calcium spike phase, but not the plateau phase occurring in response to a GnRH pulse (10 nM; 5 min) delivered immediately after pretreatment. Inhibition of the calcium spike phase was transient (recovery within 15 min) and was dependent on pretreatment concentrations of GnRH. Calcium spike phase inhibition by GnRH pretreatment prevented increased LH release from PKC-depleted cells in response to a subsequent pulse of GnRH, but not from gonadotropes with normal levels of PKC. This suggests that initial LH release is dependent on changes in [Ca2+]i, but enhancement of LH release after periods of elevated GnRH concentrations may be dependent on PKC.


Assuntos
Cálcio/metabolismo , Citosol/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante/metabolismo , Oxocinas , Animais , Células Cultivadas , Feminino , Toxinas Marinhas/farmacologia , Proteína Quinase C/fisiologia , Ratos , Ratos Endogâmicos
13.
Endocrinology ; 133(2): 496-504, 1993 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8344195

RESUMO

This study was undertaken to assess the role of protein kinase C (PKC) in activin action in the rat pituitary. Pretreatment with 500 nM phorbol 12-myristate 13-acetate (PMA) for 22-24 h reduced subsequent FSH and LH release (percentage of total cellular FSH and LH released) in response to 100 nM PMA. This action persisted for 2 days after the pretreatment. Pretreatment with 500 nM 4 alpha-phorbol 12,13-didecanoate (4 alpha PDD, a phorbol ester which does not activate PKC) did not affect cell responsiveness to 100 nM PMA. Both PKC-down-regulated cells and cells with a full complement of PKC responded similarly to 100 nM GnRH and 100 microM A23187 during this period. Incubation with 50 ng/ml activin A for 48 h significantly increased both FSH release and total FSH (extracellular plus intracellular) compared to corresponding basal values in PMA-pretreated cells, as well as in vehicle-or 4 alpha PDD-pretreated cells. Activin stimulation of basal FSH release and total FSH was significantly more potent in PMA-pretreated cells than in cells not pretreated with PMA. Activin did not alter basal LH release or total LH in vehicle- or 4 alpha PDD-pretreated cells but significantly increased both in PMA-pretreated cells. When PMA was present only during the initial 2 h of the 22- to 24-h pretreatment period at 50 nM, PKC was not down-regulated. In these cells, the potency of activin stimulation of basal FSH release was not affected, but stimulation of basal LH release by activin was still observed. These results suggest that PKC is not required for activin to stimulate FSH release but is involved as a modulator of potency and specificity of the activin action.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Inibinas/farmacologia , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Proteína Quinase C/metabolismo , Ativinas , Alcaloides/farmacologia , Animais , Calcimicina/farmacologia , Dimetil Sulfóxido/farmacologia , Feminino , Hormônio Liberador de Gonadotropina/farmacologia , Cinética , Hipófise/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Estaurosporina , Acetato de Tetradecanoilforbol/farmacologia
14.
Endocrinology ; 131(6): 2681-9, 1992 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1332845

RESUMO

After initial GnRH pretreatment (10 nM, 5 h), subsequent GnRH-stimulated LH release from the gonadotrope was diminished (1 microM GnRH stimulated release of 36.4 +/- 1.4% total cellular LH over 3 h in cells initially pretreated with medium alone compared to 27.4 +/- 1.2% in GnRH-pretreated cells); however, inositol phosphate (IP) production in response to the releasing hormone remained unaffected (1 microM GnRH provoked IP accumulation of 161 +/- 9% above basal levels after 45 min in control cells and 162 +/- 11% in GnRH-pretreated cells). Pretreatment of pituitary cell cultures with NaF (a guanyl nucleotide binding protein activator, 10 mM, 3 h) also decreased subsequent GnRH-stimulated LH release, and in addition, provoked a decrease in GnRH receptor number, an increase in GnRH receptor affinity, reduction of GnRH-stimulated IP production to basal levels, and an increase in the amount of LH released in response to stimulation with the calcium ionophore A23187. In order to determine if the changes in LH release were a result of decreased IP production and/or decreased GnRH receptor binding, the time course of recovery to control levels of these processes was assessed. GnRH receptor binding continued to decrease after NaF pretreatment, reaching a nadir (62% of control) at 6 h after the pretreatment period and recovering at 48 h (90% of control). In contrast, GnRH-provoked IP accumulation did not return to control levels even after 48 h of recovery after NaF pretreatment (1 microM GnRH-stimulated IP accumulation in NaF-pretreated cells was 57% compared to control cells after 48 h of recovery). GnRH-stimulated LH release was inhibited immediately after NaF pretreatment (1 microM GnRH-stimulated LH release in NaF-pretreated cells was 65% of control levels). Cells began to recover within 3 h (80% of control) and were almost completely recovered by 6 h (90% of control). A23187-provoked LH release was enhanced immediately after NaF pretreatment (30 microM A23187-stimulated LH release in NaF-pretreated cells was 170% of control levels). Responsiveness to ionophore was 133% of control by 0.5 h, and complete recovery was measured within 1 h (100% of control). Furthermore, both NaF and GnRH pretreatment still provoked a decrease in gonadotrope responsiveness when IP production was inhibited by the phospholipase C inhibitor U-73122. The results suggest that the development of gonadotrope desensitization (by either NaF or GnRH pretreatment) can be uncoupled from changes in IP production.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Fosfatos de Inositol/biossíntese , Hormônio Luteinizante/metabolismo , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Receptores LHRH/metabolismo , Animais , Calcimicina/farmacologia , Células Cultivadas , Tolerância a Medicamentos , Estrenos/farmacologia , Feminino , Pirrolidinonas/farmacologia , Ratos , Ratos Sprague-Dawley , Fluoreto de Sódio/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores
15.
Endocrinology ; 130(4): 2101-5, 1992 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1312442

RESUMO

The number of GnRH receptors on gonadotropes is regulated by GnRH as well as by heterologous modulators. We have used the density shift technique to measure the synthetic rate of GnRH receptors in pituitary cell cultures and found it to be stimulated by GnRH, an action that is antagonized by inhibin. In the present study, we evaluated the effects of activin-A on the GnRH receptor synthesis rate as well as effects of activin on stimulation of GnRH receptor synthesis by the homologous hormone. Recombinant human activin-A (50 ng/ml) was incubated with pituitary cell cultures from female weanling rats and the incorporation of densely labeled amino acids into receptors for GnRH was measured. The rate of GnRH receptor synthesis of cells treated with activin (50 ng/ml) together with either GnRH (0.1 ng/ml) or inhibin (12 ng/ml) was also quantified. Activin significantly stimulated the synthetic rate of GnRH receptors similarly to that observed after GnRH treatment (time for synthesis of half the population of GnRH receptors was 12.6 +/- 1.1, 16.1 +/- 1.3 vs. 28.3 +/- 1.2 h for GnRH, activin, and control, respectively), although the time course for stimulation by GnRH and activin appeared to differ. Inclusion of activin in cultures did not affect homologous stimulation of GnRH receptor synthesis. The stimulatory effects of activin were unaffected by combined treatment with inhibin (t1/2 of synthesis 17.2 +/- 2.0 h). Together, these data indicate that activin stimulates GnRH receptor synthesis in cell culture through a distinct mechanism from GnRH. Additionally, inhibin did not antagonize the stimulatory effects of activin on synthesis of GnRH receptors. This is, to our knowledge, the first demonstration of an action of activin-A on GnRH receptor synthesis.


Assuntos
Inibinas/farmacologia , Receptores LHRH/biossíntese , Ativinas , Animais , Células Cultivadas , Feminino , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Ratos , Proteínas Recombinantes/farmacologia
16.
Endocrinology ; 120(1): 160-9, 1987 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3023025

RESUMO

The coupling between GnRH-stimulated phosphoinositide (PI) turnover and LH release has been investigated in rat pituitary cell cultures. Accumulation of [3H]inositol phosphates ([3H]IPs) formed by hydrolysis of PIs was measured in cells that had been preloaded with [3H]myo-inositol. GnRH stimulated both LH release and incorporation of [3H]inositol into total [3H]IPs with similar dose and time dependencies. [3H] IP production in response to GnRH could be blocked by a GnRH antagonist, but was stimulated by a compound that provokes receptor microaggregation. GnRH-stimulated IP production persisted in the presence of either the Ca2+ channel blocker D600 or the calmodulin antagonist pimozide at concentrations that reduced LH release to 60% and 20% of control, respectively. Stimulated [3H]IP production was inhibited at higher concentrations of D600. In 1-h incubations, GnRH-stimulated [3H]IP production, but not LH release, was markedly inhibited by the protein kinase C activators phorbol myristate acetate and 1,2-dioctanoylglycerol. These findings indicate that in the gonadotrope, GnRH-stimulated LH release and [3H]IP production are closely coupled to receptor activation by an agonist; Ca2+ antagonists uncouple stimulated LH release from [3H]IP production; and protein kinase C activators uncouple stimulated PI turnover from LH release. Thus, GnRH-stimulated production of PI metabolites, as measured by [3H]IP accumulation, is apparently not sufficient to support LH release in the absence of Ca2+. In addition, GnRH-stimulated LH release is apparently not dependent on full expression of the PI response.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Fosfatos de Inositol/fisiologia , Hormônio Luteinizante/metabolismo , Adeno-Hipófise/metabolismo , Fosfatos Açúcares/fisiologia , Animais , Cálcio/fisiologia , Calmodulina/fisiologia , Diglicerídeos/farmacologia , Feminino , Galopamil/farmacologia , Fosfatidilinositóis/metabolismo , Pimozida/farmacologia , Adeno-Hipófise/fisiologia , Proteína Quinase C/metabolismo , Ratos , Receptores LHRH/fisiologia , Acetato de Tetradecanoilforbol/farmacologia
17.
Endocrinology ; 120(1): 222-9, 1987 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3023028

RESUMO

Exposure of pituitary cell cultures to GnRH causes gonadotropin release, receptor capping, internalization, and loss as well as altered responsiveness of the target cell. In the present study, the relationship between loss of gonadotrope secretory responsiveness to GnRH (desensitization) and internalization of the GnRH-receptor complex was examined. Pituitary cell cultures were pretreated (30 min) with vinblastine (100 microM, a concentration that prevents measurable receptor internalization) or with medium containing carrier only, incubated with 10(-7) M GnRH (a desensitizing concentration) with or without vinblastine or with medium alone for 60 min, and finally washed and rechallenged for 3 h with increasing concentrations of GnRH to assess the degree of desensitization as determined by LH release. Results indicate that vinblastine had no measurable effect on the ability of GnRH to stimulate LH release or desensitize the cells. In a second series of studies, a GnRH analog (D-Lys6-GnRH) was immobilized to a cross-linked agarose matrix. The covalent link was shown to be stable by biological, immunological, and physical criteria. This product bound to the GnRH receptor and provoked LH release, but was not internalized, as determined by GnRH receptor binding assays. Cultured cells were treated with either 10(-9) M free analog or an equivalent concentration of coupled analog (as measured by LH release) for 3 h. Cells were washed, then rechallenged with GnRH to assess desensitization. Both the free and coupled analogs provoked an equivalent degree of desensitization. While a significant degree of desensitization also occurred in the presence of 3 mM EGTA (conditions that totally inhibited GnRH-stimulated LH release), the loss of responsiveness was not as great as in the absence of EGTA, indicating that partial depletion of available LH may play a role in GnRH-stimulated gonadotrope desensitization. The present findings suggest that GnRH receptor internalization and LH release can be uncoupled and that loss of the GnRH receptor by internalization is not a sufficient explanation for GnRH-mediated desensitization of the gonadotrope.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante/metabolismo , Adeno-Hipófise/fisiologia , Receptores LHRH/fisiologia , Animais , Células Cultivadas , Endocitose , Feminino , Hormônios/farmacologia , Técnicas In Vitro , Ratos , Taxa Secretória , Vimblastina/farmacologia
18.
Endocrinology ; 137(8): 3602-5, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8754794

RESUMO

Gonadotropin releasing hormone (GnRH) mediates interactions between the neural and the endocrine systems. The GnRH receptor is a member of the ubiquitous seven-transmembrane segment G-protein-coupled receptor class and is the target of drug development for treatment of breast and prostate cancer, regulation of fertility, endometriosis and a range of other medical and veterinary uses. This study shows that occupancy of the receptor by an agonist (but not an antagonist) promotes receptor-receptor interactions which appears to be an early event in hormone action.


Assuntos
Hormônio Liberador de Gonadotropina/agonistas , Receptores LHRH/efeitos dos fármacos , Transdução de Sinais/fisiologia , Animais , Bovinos , Feminino , Hormônio Liberador de Gonadotropina/farmacologia , Lactoperoxidase/farmacologia , Hipófise/citologia , Hipófise/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores LHRH/fisiologia
19.
Endocrinology ; 112(1): 408-10, 1983 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-6401180

RESUMO

In the present work we describe homologous desensitization of gonadotropin release from pituitary cells in response to short-term exposure to GnRH. Like the release process itself, desensitization requires receptor occupancy by an agonist. In contrast, however, inhibition of LH release by chelation of extracellular calcium or blockade of the calcium ion channel does not inhibit desensitization. Further, stimulation of gonadotropin release by ionophore A23187 (which mobilizes calcium without GnRH receptor occupancy) does not lead to desensitization. These findings indicate that the decrease in pituitary sensitivity to GnRH due to an initial exposure to the peptide can be uncoupled from LH release. The data provide the first evidence of a calcium-independent biological effect of the releasing hormone in cultured pituitary cells.


Assuntos
Cálcio/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Hipófise/efeitos dos fármacos , Animais , Calcimicina/farmacologia , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Ratos , Ratos Endogâmicos , Fatores de Tempo
20.
Endocrinology ; 109(6): 2040-5, 1981 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-6273126

RESUMO

Three different approaches are described which provide independent and new evidence that gonadotropin-releasing hormone (GnRH) internalization and GnRH-stimulated LH release are distinct actions of the releasing hormone. 1) Removal of GnRH from medium bathing the pituitary cell cultures resulted in the prompt return of LH release to basal levels. This finding indicated that a continuous supply of externally applied GnRH is required for the stimulation of LH release. 2) Covalent immobilization of D-Lys6-des-Gly10-Pro9-ethylamide GnRH (a GnRH agonist) on agarose beads resulted in a derivative which stimulated LH release with full efficacy. At concentrations of immobilized releasing hormone analog sufficient to evoke gonadotropin release, the quantity of LH release was restricted by the number of beads added. This finding was interpreted as evidence that the attachment of immobilized agonist was stable during the bioassay and indicated that LH release could be stimulated with full efficacy without the requirement for GnRH internalization. 3) Comparative studies using image-intensified microscopy and the cell culture bioassay showed that 100 microM vinblastin markedly inhibited large scale patching and capping of the GnRH receptor (viewed by image-intensified microscopy), but did not alter the EC50 or efficacy of LH release stimulated by GnRH or the agonist described above. These observations indicated that internalization as well as large scale patching and capping of the GnRH receptor are not required for LH release.


Assuntos
Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/fisiologia , Hormônio Luteinizante/metabolismo , Hipófise/fisiologia , Receptores de Superfície Celular/fisiologia , Animais , Células Cultivadas , Cromatografia de Afinidade , Feminino , Hormônio Liberador de Gonadotropina/síntese química , Hormônio Liberador de Gonadotropina/farmacologia , Hipófise/efeitos dos fármacos , Ácido Pirrolidonocarboxílico/análogos & derivados , Ratos , Receptores de Superfície Celular/efeitos dos fármacos , Receptores LHRH , Vimblastina/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa