Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Reproduction ; 160(2): 239-245, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32422603

RESUMO

Ovarian hyperstimulation syndrome (OHSS) is a common complication of ovarian stimulation associated with the administration of human chorionic gonadotropin (hCG) during assisted reproduction. We have determined the expression of luteinizing hormone receptor (Lhcgr) mRNA, vascular endothelial growth factor (VEGF), and its transcription factor, HIF1α, during the periovulatory period in a rodent model of OHSS and compared these results with normal ovulatory periods. These results showed that the downregulation of Lhcgr mRNA in response to conditions that mimic preovulatory LH surge was significantly impaired in the OHSS group compared to the complete downregulation seen in the control group. Most importantly, the downregulation of luteinizing hormone receptor mRNA expression following hCG administration was sustained in the control group up to 48 h, whereas it remained at significantly higher levels in the OHSS group. This impairment of hCG-induced Lhcgr downregulation in the OHSS group was accompanied by significantly elevated levels of VEGF and its transcription factor, HIF1α. Furthermore, the downregulation of Lhcgr that occurs in response to a preovulatory LH surge in normal cycles was accompanied by low levels of VEGF. This study shows that, while downregulation of Lhcgr as well as low VEGF levels are seen in response to a preovulatory LH surge in normal ovarian cycle, impaired Lhcgr downregulation and elevated VEGF levels were found in the OHSS group.


Assuntos
Gonadotropina Coriônica/farmacologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Síndrome de Hiperestimulação Ovariana/patologia , Indução da Ovulação/métodos , Receptores do LH/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Feminino , Síndrome de Hiperestimulação Ovariana/tratamento farmacológico , Síndrome de Hiperestimulação Ovariana/genética , Síndrome de Hiperestimulação Ovariana/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores do LH/genética , Substâncias para o Controle da Reprodução/farmacologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética
2.
Endocrinology ; 159(5): 2075-2082, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29579170

RESUMO

Luteinizing hormone/chorionic gonadotropin receptor (LHR) expression in the ovary is regulated by a messenger RNA (mRNA) binding protein, which specifically binds to the coding region of LHR mRNA. We have shown that miR-122, a short noncoding RNA, mediates LHR mRNA levels by modulating the expression of LHR mRNA-binding protein (LRBP) through the regulation of sterol regulatory element binding protein (SREBP) activation. The present results show that miR-122 regulates LRBP levels by increasing the processing of SREBP through the degradation of Insig1, the anchoring protein of SREBP. We present evidence showing that mRNA and protein levels of Insig1 undergo a time-dependent increase following the treatment of rat granulosa cells with follicle-stimulating hormone (FSH), which leads to a decrease in LRBP levels. Furthermore, overexpression of miR-122 using an adenoviral vector (AdmiR-122) abolished FSH-induced increases in Insig1 mRNA and protein. We further confirmed the role of Insig1 by showing that inhibition of Insig1 using a specific small interfering RNA prior to FSH treatment resulted in the abrogation of LHR upregulation. Silencing of Insig1 also reversed FSH-mediated decreases in SREBP and LRBP activation. These results show that decreased levels of miR-122 increase Insig1 and suppress SREBP processing in response to FSH stimulation of rat granulosa cells.


Assuntos
Células da Granulosa/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , MicroRNAs/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Receptores do LH/genética , Proteínas de Ligação a Elemento Regulador de Esterol/genética , Animais , Feminino , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica , Hormônios/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool) , RNA Interferente Pequeno , Proteínas de Ligação a RNA/metabolismo , Ratos , Receptores do LH/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo
3.
Endocrinology ; 158(8): 2672-2679, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28605466

RESUMO

We have shown that the transient changes in the expression of luteinizing hormone/choriogonadotropin receptor (LHCGR) messenger RNA (mRNA) during the ovarian cycle occurs, at least in part, through a posttranscriptional mechanism involving an LHCGR mRNA-binding protein (LRBP). Eukaryotic initiation factor 5A (eIF5A), an LRBP-interacting protein, participates in this process. eIF5A undergoes hypusination, a unique posttranslational modification that is necessary for its functions. This study examined the role of eIF5A in follicle-stimulating hormone (FSH)-induced LHCGR expression during follicular growth. Treatment of primary cultures of rat granulosa cells with FSH and 17ß-estradiol (E2) showed a time-dependent increase in LHCGR mRNA expression. Conversely, inhibition of endogenous hypusination of eIF5A using N1-guanyl-1,7-diaminoheptane (GC7), a hypusination inhibitor, showed a greater increase in LHCGR mRNA expression over that produced by FSH and E2 alone. Further studies were carried out to determine the mechanism by which inhibition of hypusination of eIF5A causes an increase in LHCGR mRNA expression. Because LHCGR expression is negatively regulated by LRBP, the effect of inhibiting hypusination of eIF5A on LRBP expression was examined. The results showed a decrease in the expression of LRBP mRNA and protein when hypusination of eIF5A was inhibited by GC7. Because LRBP promotes LHCGR mRNA degradation, the results of this study support the notion that by inhibiting eIF5A hypusination, FSH reduces the expression of LRBP. This increases LHCGR mRNA expression by abrogating the inhibitory action of LRBP.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Fatores de Iniciação de Peptídeos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores do LH/metabolismo , Animais , Feminino , Regulação da Expressão Gênica/fisiologia , Células da Granulosa/fisiologia , Fatores de Iniciação de Peptídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Ratos , Ratos Sprague-Dawley , Receptores do LH/genética , Fator de Iniciação de Tradução Eucariótico 5A
4.
Mol Cell Endocrinol ; 442: 81-89, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27940300

RESUMO

We have previously reported that LHCGR expression in the ovary is regulated through a post-transcriptional mechanism involving an mRNA binding protein designated as LRBP, which is regulated, at least in part, by a non-coding RNA, miR-122. Our present study examined the regulatory role of miR-122 in FSH-induced LHCGR expression during follicle development. Treatment of rat granulosa cells concurrently with FSH and 17ß estradiol showed, as expected, a time-dependent increase in LHCGR mRNA levels as well as hCG-induced progesterone production. However, miR-122 expression was decreased during the early time periods, which preceded the increased expression of LHCGR mRNA. The role of miR-122 in FSH-induced LHCGR mRNA expression was then examined by overexpressing miR-122 prior to FSH stimulation by infecting granulosa cells with an adenoviral vector containing a miR-122 insert (AdmiR-122). Pretreatment with AdmiR-122 resulted in complete abrogation of FSH- mediated upregulation of LHCGR. AdmiR-122 also blocked FSH-induced decrease in LRBP expression and increased the binding of LHCGR mRNA to LRBP. Based on these results, we conclude that miR-122 plays a regulatory role in LHCGR expression by modulating LRBP levels during FSH-induced follicle growth.


Assuntos
MicroRNAs/metabolismo , Folículo Ovariano/metabolismo , Ovário/metabolismo , Receptores do LH/metabolismo , Animais , Estradiol/metabolismo , Feminino , Hormônio Foliculoestimulante/metabolismo , Células da Granulosa/metabolismo , Progesterona/metabolismo , RNA Mensageiro/metabolismo , Ratos , Regulação para Cima/fisiologia
5.
Endocrinology ; 156(9): 3370-80, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26125464

RESUMO

LH/human chorionic gonadotropin receptor (LHR) undergoes down-regulation during preovulatory LH surge or in response to exposure to a supraphysiological concentration of its ligands through a posttranscriptional mechanism involving an RNA binding protein designated as LHR mRNA binding protein (LRBP). miR-122, a short noncoding RNA, has been shown to mediate the up-regulation of LRBP. In the present study, we show that inhibition of miR-122 using a locked nucleic acid (LNA)-conjugated antagomir suppressed human chorionic gonadotropin (hCG)-induced up-regulation of LRBP as well as its association with LHR mRNA, as analyzed by RNA EMSA. Most importantly, inhibition of miR-122 resulted in the abolishment of hCG-mediated LHR mRNA down-regulation. We also show that the transcription factor, sterol regulatory element binding protein (SREBP) (SREBP-1a and SREBP-2 isoforms), is an intermediate in miR-122-mediated LHR mRNA regulation. HCG-stimulated increase in the activation of both SREBP-1a and SREBP-2 was inhibited by pretreatment with the miR-122 antagomir. The inhibition of cAMP/protein kinase A (PKA) and ERK pathways, upstream activators of miR-122, abolished SREBP activation after hCG treatment. SREBP-mediated regulation of LRBP expression is mediated by recruitment of LRBP promoter element to SREBP-1a, because chromatin immunoprecipitation assay revealed that association of LRBP promoter to SREBP was increased by hCG treatment. Pretreatment with miR-122 antagomir suppressed this response. Inhibition of SREBP activation by pretreating the rats with a chemical compound, fatostatin abrogated hCG-induced up-regulation of LRBP mRNA and protein. Fatostatin also inhibited LHR-LRBP mRNA-protein complex formation and LHR down-regulation. These results conclusively show that miR-122 plays a regulatory role in LH/hCG-induced LHR mRNA down-regulation by increasing LRBP expression through the activation of SREBP pathway.


Assuntos
MicroRNAs/metabolismo , Ovário/metabolismo , Receptores do LH/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Animais , Antagomirs , Gonadotropina Coriônica , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Regulação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Oligonucleotídeos , Fosfotransferases (Aceptor do Grupo Álcool) , Piridinas , Proteínas de Ligação a RNA/metabolismo , Ratos Sprague-Dawley , Ribonucleoproteínas/metabolismo , Tiazóis
6.
Mol Cell Endocrinol ; 413: 90-5, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26116232

RESUMO

Luteinizing hormone receptor (LHR) mRNA expression in the ovary is regulated post-transcriptionally by an LH receptor mRNA binding protein (LRBP). Eukaryotic initiation factor 5A (EIF5A), identified as an LRBP-interacting protein plays a crucial role in LHR mRNA expression. In this study, we have demonstrated that during hCG-induced LHR downregulation, a significant upregulation of eIF5A mRNA expression and hypusination of eIF5A protein occurs in a time dependent manner. Pretreatment with H89, a specific inhibitor of PKA, and U0126, a specific inhibitor of ERK1/2 significantly inhibited both hCG-induced eIF5A mRNA expression and hypusination of eIF5A protein. Pretreatment with GC7, a specific inhibitor of eIF5A hypusination significantly abolished hCG-induced LRBP mRNA and protein expression. Furthermore, GC7 pretreatment significantly inhibited hCG-induced interaction of LRBP with LHR mRNA as assessed by RNA electrophoretic mobility gel shift assay (REMSA). GC7 treatment also reversed LHR mRNA downregulation. Taken together, these results suggest that hCG-induced LHR mRNA downregulation is mediated by cAMP-PKA-ERK1/2 signaling leading to activation of eIF5A hypusination.


Assuntos
AMP Cíclico/metabolismo , Regulação da Expressão Gênica/fisiologia , Lisina/análogos & derivados , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Ovário/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Receptores do LH/biossíntese , Animais , Proteínas Quinases Dependentes de AMP Cíclico , Feminino , Hormônio Luteinizante/metabolismo , Lisina/metabolismo , Fatores de Iniciação de Peptídeos , Fosfotransferases (Aceptor do Grupo Álcool) , RNA Mensageiro/biossíntese , Proteínas de Ligação a RNA/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Iniciação de Tradução Eucariótico 5A
7.
Indian J Med Res ; 140 Suppl: S112-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25673531

RESUMO

A specific luteinizing hormone receptor (LHR) mRNA binding protein (LRBP) has been identified and purified. This LH receptor mRNA binding protein selectively binds to the polypyrimidine rich bipartite sequence in the coding region of the LHR mRNA and accelerates its degradation. In response to preovulatory LH surge, the LH receptor expression in the ovary undergoes downregulation by accelerated degradation of LH receptor mRNA through the involvement of this RNA binding protein. Here we describe the intracellular mechanism triggered by LH/hCG (human chorionic gonadotropin) that leads to the regulated degradation of LH receptor mRNA. Downregulation of LH receptor mRNA was induced by treatment of cultured human granulosa cells with 10 IU of hCG. Activation of downstream target, extracellular signal-regulated protein kinase 1 and 2 (ERK 1/2) showed an increase within five min and sustained up to 1 h. Confocal analysis showed that ERK1/2 translocates to the nucleus after 15 min of hCG treatment. This leads to an increase in LRBP expression which then causes downregulation of LH receptor mRNA by accelerating its degradation. Treatment with UO126 or transfection with ERK specific siRNA (small interfering RNA) resulted in the abolishment of ERK activation as well as LHR mRNA downregulation. RNA electrophoretic mobility gel shift assay of the cytosolic fractions showed that hCG-induced increase in the LH receptor mRNA binding activity was also abrogated by these treatments. These results show that LH/hCG-induced LH receptor mRNA downregulation is initiated by the activation of ERK1/2 pathway by regulating the expression and activity of LH receptor mRNA binding activity.


Assuntos
Hormônio Luteinizante/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Ovário/metabolismo , Proteólise , Proteínas de Ligação a RNA/metabolismo , Receptores do LH/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Northern Blotting , Butadienos , Células Cultivadas , Gonadotropina Coriônica/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Células da Granulosa/metabolismo , Humanos , Nitrilas , Ovário/citologia , RNA Interferente Pequeno/farmacologia
8.
Endocrinology ; 154(12): 4826-34, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24064360

RESUMO

LH receptor (LHR) expression in the ovary is regulated by the RNA binding protein, (LHR mRNA binding protein [LRBP]), which has been identified as being mevalonate kinase. This study examined the role of microRNA miR-122 in LRBP-mediated LHR mRNA expression. Real-time PCR analysis of ovaries from pregnant mare serum gonadotropin/human chorionic gonadotropin (hCG)-primed female rats treated with hCG to down-regulate LHR expression showed that an increase in miR-122 expression preceded LHR mRNA down-regulation. The expression of miR-122 and its regulation was confirmed using fluorescent in situ hybridization of the frozen ovary sections using 5'-fluorescein isothiocyanate-labeled miR-122 locked nucleic acid probe. The increased expression of miR-122 preceded increased expression of LRBP mRNA and protein, and these increases were followed by LHR mRNA down-regulation. Inhibition of protein kinase A (PKA) and ERK1/2 signaling pathways by H89 and UO126, respectively, attenuated the hCG-mediated up-regulation of miR-122 levels. This was also confirmed in vitro using human granulosa cells. These results suggest the possibility that hCG-mediated miR-122 expression is mediated by the activation of cAMP/PKA/ERK signaling pathways. Inhibition of miR-122 by injection of the locked nucleic acid-conjugated antagomir of miR-122 abrogated the hCG-mediated increases in LRBP protein expression. Because it has been previously shown that miR-122 regulates sterol regulatory element-binding proteins (SREBPs) and SREBPs, in turn, regulate LRBP expression, the role of SREBPs in miR-122-mediated increase in LRBP expression was then examined. The levels of active forms of both SREBP-1a and SREBP-2 were increased in response to hCG treatment, and the stimulatory effect was sustained up to 4 hours. Taken together, our results suggest that hCG-induced down-regulation of LHR mRNA expression is mediated by activation of cAMP/PKA/ERK pathways to increase miR-122 expression, which then increases LRBP expression through the activation of SREBPs.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , MicroRNAs/metabolismo , Ovário/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Células Cultivadas , Gonadotropina Coriônica/farmacologia , AMP Cíclico/genética , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Hormônio Luteinizante/farmacologia , Sistema de Sinalização das MAP Quinases/fisiologia , MicroRNAs/genética , Ovário/citologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Ratos , Ratos Sprague-Dawley , Proteínas de Ligação a Elemento Regulador de Esterol/genética , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Regulação para Cima
9.
Mol Cell Endocrinol ; 366(1): 81-9, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23261705

RESUMO

The present study examined the effect of insulin-mediated activation of the mammalian target of rapamycin complex 1 (MTORC1) signaling network on the proliferation of primary culture of theca-interstitial (T-I) cells. Our results show that insulin treatment increased proliferation of the T-I cells through the MTORC1-dependent signaling pathway by increasing cell cycle regulatory proteins. Inhibition of ERK1/2 signaling caused partial reduction of insulin-induced phosphorylation of RPS6KB1 and RPS6 whereas inhibition of PI3-kinase signaling completely blocked the insulin response. Pharmacological inhibition of MTORC1 with rapamycin abrogated the insulin-induced phosphorylation of EIF4EBP1, RPS6KB1 and its downstream effector, RPS6. These results were further confirmed by demonstrating that knockdown of Mtor using siRNA reduced the insulin-stimulated MTORC1 signaling. Furthermore, insulin-stimulated T-I cell proliferation and the expression of cell cycle regulatory proteins CDK4, CCND3 and PCNA were also blocked by rapamycin. Taken together, the present studies show that insulin stimulates cell proliferation and cell cycle regulatory proteins in T-I cells via activation of the MTORC1 signaling pathway.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Insulina/farmacologia , Complexos Multiproteicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Células Tecais/citologia , Animais , Proteínas de Transporte/metabolismo , Bovinos , Proliferação de Células/efeitos dos fármacos , Feminino , Inativação Gênica/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular , Alvo Mecanístico do Complexo 1 de Rapamicina , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Complexos Multiproteicos/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína S6 Ribossômica/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Fatores de Tempo
10.
Fertil Steril ; 98(1): 207-14, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22608319

RESUMO

OBJECTIVE: To determine whether metformin has direct effects on ovarian theca-interstitial (T-I) cell proliferation through activation of adenosine monophosphate-activated protein kinase (AMPK). DESIGN: In vitro experimental study. SETTING: Academic medical center laboratory. ANIMAL(S): Immature Sprague-Dawley female rats. INTERVENTION(S): Ovarian T-I cells were isolated, purified, and cultured in the absence (control) or presence of insulin (1 µg/mL) with or without metformin or other activators/inhibitors of AMPK (AICAR, compound C). MAIN OUTCOME MEASURE(S): Proliferation assessed by determination of expression levels of proteins involved in cell cycle progression, cyclin D3, and cyclin-dependent kinase 4 (CDK4) with Western blot analysis, and determination of DNA synthesis with bromodeoxyuridine (BrdU) incorporation assay; activation of AMPK, Erk1/2, and S6K1 determined by Western blot analysis with the use of antibodies specific for the phosphorylated (activated) forms. RESULT(S): Metformin inhibited insulin-induced ovarian T-I cell proliferation and the up-regulation of the cell cycle regulatory proteins, cyclin D3 and CDK4. Metformin independently activated AMPK in a dose-dependent manner. Treatment with metformin inhibited insulin-induced activation of Erk1/2 and S6K1. This effect was reversed with the addition of compound C, a known AMPK inhibitor. CONCLUSION(S): Metformin directly inhibits proliferation of ovarian T-I cells via an AMPK-dependent mechanism. These findings further validate the potential benefits of metformin in the treatment of conditions associated with hyperinsulinemia and excessive growth of ovarian T-I cells (such as polycystic ovary syndrome).


Assuntos
Proliferação de Células/efeitos dos fármacos , Metformina/farmacologia , Ovário/efeitos dos fármacos , Células Tecais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/fisiologia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Feminino , Hipoglicemiantes/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ovário/citologia , Ratos , Ratos Sprague-Dawley , Proteínas Quinases S6 Ribossômicas/metabolismo , Células Tecais/citologia , Células Tecais/fisiologia
11.
Endocrinology ; 153(6): 2831-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22454147

RESUMO

We have previously reported that 5α-dihydrotestosterone (DHT) inhibits FSH-mediated granulosa cell proliferation by reducing cyclin D2 mRNA expression and blocking cell cycle progression at G1/S phase. The present study investigated the role of AMP activated protein kinase (AMPK) in DHT-mediated inhibition of granulosa cell proliferation. Granulosa cells harvested from 3-d estradiol primed immature rats were exposed to different concentrations of DHT (0, 45, and 90 ng/ml) for 24 h. Western blot analysis of immunoprecipitated AMPK showed a dose-dependent activation (P < 0.05) as evidenced by the increased phosphorylation at thr 172. In addition, time-courses studies (0, 6, 12, and 24 h) using DHT (90 ng/ml) showed a time-dependent increase in AMPK activation with maximum effect at 24 h. FSH inhibited AMPK phosphorylation and promoted granulosa cell proliferation, but pretreatment with DHT (90 ng/ml) for 24 h prior to FSH treatment reduced this effect. Pharmacological activation of AMPK with 5-aminoimidazole-4-carboxamide-1-ß4-ribofuranoside abolished FSH-mediated ERK phosphorylation, indicating that AMPK is a negative upstream regulator of ERK. Furthermore, inhibition of AMPK activation by compound C reversed the DHT-mediated reduction in positive cell cycle regulator, cyclin D2, and 5-bromo-2'-deoxyuridine incorporation. These results suggest that elevated levels of DHT activate AMPK, which in turn inhibits ERK phosphorylation. Thus, inhibition of ERK phosphorylation by activated AMPK in response to DHT might contribute to decreased granulosa cell mitogenesis and ovulatory dysfunction seen in hyperandrogenic states.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proliferação de Células/efeitos dos fármacos , Di-Hidrotestosterona/farmacologia , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Androgênios/farmacologia , Animais , Western Blotting , Células Cultivadas , Ciclina D2/genética , Ciclina D2/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Hormônios/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
12.
Mol Endocrinol ; 25(2): 282-90, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21147848

RESUMO

The ligand-induced down-regulation of LH receptor (LHR) expression in the ovaries, at least in part, is regulated by a posttranscriptional process mediated by a specific LH receptor mRNA binding protein (LRBP). The LH-mediated signaling pathways involved in this process were examined in primary cultures of human granulosa cells. Treatment with 10 IU human chorionic gonadotropin (hCG) for 12 h resulted in the down-regulation of LHR mRNA expression while producing an increase in LHR mRNA binding to LRBP as well as a 2-fold increase in LRBP levels. The activation of ERK1/2 pathway in LH-mediated LHR mRNA down-regulation was also established by demonstrating the translocation of ERK1/2 from the cytosol to the nucleus using confocal microcopy. Inhibition of protein kinase A using H-89 or ERK1/2 by U0126 abolished the LH-induced LHR mRNA down-regulation. These treatments also abrogated both the increases in LRBP levels as well as the LHR mRNA binding activity. The abolishment of the hCG-induced increase in LRBP levels and LHR mRNA binding activity was further confirmed by transfecting granulosa cells with ERK1/2 specific small interfering RNA. This treatment also reversed the hCG-induced down-regulation of LHR mRNA. These data show that LH-regulated ERK1/2 signaling is required for the LRBP-mediated down-regulation of LHR mRNA.


Assuntos
Regulação para Baixo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , RNA Mensageiro/genética , Receptores do LH/genética , Western Blotting , Butadienos/farmacologia , Linhagem Celular , Núcleo Celular/metabolismo , Gonadotropina Coriônica/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/genética , Feminino , Imunofluorescência , Células da Granulosa/metabolismo , Humanos , Isoquinolinas/farmacologia , Microscopia Confocal , Nitrilas/farmacologia , Ovário , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Reação em Cadeia da Polimerase , Processamento Pós-Transcricional do RNA , RNA Interferente Pequeno/farmacologia , Receptores do LH/metabolismo , Transdução de Sinais , Sulfonamidas/farmacologia
13.
Endocrine ; 37(1): 187-93, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20963569

RESUMO

The objective of the study was to evaluate the effect of valproic acid (VPA) on ovarian androgen biosynthesis in primary cultures of theca-interstitial (T-I) cells isolated from rat ovaries. Ovarian T-I cells were cultured with VPA in the presence or absence of hCG. VPA did not increase basal or hCG-stimulated androgen synthesis when added to primary cultures of T-I cells. However, the addition of VPA caused a marked concentration-dependent inhibitory effect on hCG-stimulated androstendione synthesis. Treatment of T-I cells with 8-Bromo-cAMP resulted in a marked increase in the production of androstenedione, and VPA inhibited this stimulatory effect, suggesting that the mechanism of VPA's inhibitory effect on androstenedione production occurs at a step after second messenger activation. Treatment of T-I cells with hCG resulted in a significant increase in the mRNA expression of steroidogenic enzymes CYP17A1 and 17ß-hydroxysteroid dehydrogenase. Addition of VPA sharply blunted the stimulatory effect of hCG, reducing the mRNA expression of the steroidogenic enzymes to basal levels. In conclusion, VPA exerts an inhibitory effect on hCG-stimulated androgen synthesis in rat T-I cells.


Assuntos
Androgênios/metabolismo , Antimaníacos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Hormônios Esteroides Gonadais/metabolismo , Células Tecais/efeitos dos fármacos , Células Tecais/metabolismo , Ácido Valproico/farmacologia , 17-Hidroxiesteroide Desidrogenases/genética , 17-Hidroxiesteroide Desidrogenases/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/antagonistas & inibidores , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Androstenodiona/metabolismo , Animais , Antimaníacos/efeitos adversos , Células Cultivadas , Gonadotropina Coriônica/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Concentração Osmolar , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Esteroide 17-alfa-Hidroxilase/genética , Esteroide 17-alfa-Hidroxilase/metabolismo , Ácido Valproico/efeitos adversos
14.
Endocrinology ; 151(10): 5030-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20810561

RESUMO

Elevated levels of 5α-reduced androgens have been shown to be associated with hyperandrogenism and hyperinsulinemia, the leading causes of ovulatory dysfunction in women. 5α-Dihydrotestosterone reduces ovarian granulosa cell proliferation by inhibiting FSH-mediated mitogenic signaling pathways. The present study examined the effect of insulin on 5α-reductase, the enzyme that catalyses the conversion of androgens to their 5α-derivatives. Granulosa cells isolated from immature rat ovaries were cultured in serum-free, phenol red-free DMEM-F12 media and treated with different doses of insulin (0, 0.1, 1.0, and 10.0 µg/ml) for different time intervals up to 12 h. The expression of 5α-reductase type 1 mRNA, the predominant isoform found in granulosa cells, showed a significant (P<0.05) increase in response to the insulin treatment up to 12 h compared with control. The catalytic activity of 5α-reductase enzyme was also stimulated in a dose-depended manner (P<0.05). Inhibiting the Akt-dependent signaling pathway abolished the insulin-mediated increase in 5α-reductase mRNA expression, whereas inhibition of the ERK-dependent pathway had no effect. The dose-dependent increase in 5α-reductase mRNA expression as well as catalytic activity seen in response to insulin treatment was also demonstrated in the human granulosa cell line (KGN). In addition to increased mRNA expression, a dose-dependent increase in 5α-reductase protein expression in response to insulin was also seen in KGN cells, which corroborated well with that of mRNA expression. These results suggest that elevated levels of 5α-reduced androgens seen in hyperinsulinemic conditions might be explained on the basis of a stimulatory effect of insulin on 5α-reductase in granulosa cells. The elevated levels of these metabolites, in turn, might adversely affect growth and proliferation of granulosa cells, thereby impairing follicle growth and ovulation.


Assuntos
3-Oxo-5-alfa-Esteroide 4-Desidrogenase/genética , Células da Granulosa/efeitos dos fármacos , Insulina/farmacologia , Proteínas de Membrana/genética , Proteína Oncogênica v-akt/fisiologia , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/enzimologia , Células da Granulosa/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Proteína Oncogênica v-akt/metabolismo , Ovário/efeitos dos fármacos , Ovário/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Regulação para Cima/efeitos dos fármacos
15.
Mol Endocrinol ; 24(9): 1782-93, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20660299

RESUMO

In addition to playing a cardinal role in androgen production, LH also regulates growth and proliferation of theca-interstitial (T-I) cells. Here, we show for the first time that LH/human chorionic gonadotropin (hCG) regulates T-I cell proliferation via the mammalian target of rapamycin complex 1 (mTORC1) signaling network. LH/hCG treatment showed a time-dependent stimulation of T-I cell proliferation and phosphorylation of protein kinase B (AKT), ERK1/2, and ribosomal protein (rp)S6 kinase 1 (S6K1), and its downstream effector, rpS6. Pharmacological inhibition of ERK1/2 signaling did not block the hCG-induced phosphorylation of tuberin, the upstream regulator of mTORC1 or S6K1, the downstream target of mTORC1. However, inhibition of AKT signaling completely blocked the hCG response. Furthermore, the AKT-specific inhibitor abolished forskolin (FSK)-stimulated phosphorylation of AKT, tuberin, S6K1, and rpS6. Human CG and FSK-mediated phosphorylation of AKT and downstream targets of mTORC1 were attenuated by inhibition of adenylyl cyclase. Pharmacologic targeting of mTORC1 with rapamycin also abrogated hCG or FSK-induced phosphorylation of S6K1, rpS6, and eukaryotic initiation factor 4E binding protein 1. In addition, hCG or FSK-mediated up-regulation of the cell cycle regulatory proteins cyclin-dependent kinase 4, cyclin D3, and proliferating cell nuclear antigen was blocked by rapamycin. These results were further confirmed by demonstrating that knockdown of mTORC1 using small interfering RNA abolished hCG-mediated increases in cell proliferation and the expression of cyclin D3 and proliferating cell nuclear antigen. Taken together, the present studies show a novel intracellular signaling pathway for T-I cell proliferation involving LH/hCG-mediated activation of the AKT/mTORC1 signaling cascade.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Gonadotropina Coriônica/farmacologia , AMP Cíclico/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tecais/citologia , Fatores de Transcrição/metabolismo , Animais , Proteínas de Transporte/metabolismo , Proliferação de Células/efeitos dos fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina D3/genética , Ciclina D3/metabolismo , Ativação Enzimática/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Proteína S6 Ribossômica/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Sirolimo/farmacologia , Células Tecais/efeitos dos fármacos , Células Tecais/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/metabolismo
16.
Mol Cell Endocrinol ; 328(1-2): 28-33, 2010 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-20619315

RESUMO

Vascular endothelial growth factor A (VEGF-A) is one of the most important regulators of ovarian angiogenesis. In this study, we examined the temporal relationship between VEGF-A and luteinizing hormone receptor (LHR) mRNA expression during ligand-induced down-regulation of LHR. Immature female rats were treated with pregnant mare's serum gonadotropin followed by 25 IU hCG 56 h later (day 0). On day 5, treatment with hCG (50 IU) to down-regulate LHR showed a temporal decrease in VEGF-A mRNA and protein levels in parallel with decreasing LHR mRNA. This effect was specific since the expression of CYP11A1 mRNA showed no decline. Examination of VEGF-A mRNA expression, using in situ hybridization histochemistry with (35)S-labeled antisense VEGF-A mRNA probe, showed intense signal in the corpora lutea on day 5. Treatment with 50 IU hCG to down-regulate LHR mRNA showed a decline in the intensity of VEGF-A mRNA in the corpora lutea. VEGF-A mRNA expression returned to control level 53 h later when the expression of LHR mRNA also recovered. These results show that the transient down-regulation of VEGF-A mRNA and protein closely parallels the ligand-induced down-regulation of LHR mRNA. The present study establishes a close association between VEGF-A and LHR mRNA expression, suggesting the possibility that VEGF-A-induced vascularization of the ovary is dictated by the expression of LHR and this might play a regulatory role in ovarian physiology.


Assuntos
Hormônio Luteinizante/farmacologia , Ovário/efeitos dos fármacos , Receptores do LH/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Separação Celular , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Regulação para Baixo/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Ligantes , Células Lúteas/citologia , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Ovário/irrigação sanguínea , Ovário/metabolismo , Ovário/fisiologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores do LH/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Mol Cell Endocrinol ; 329(1-2): 26-32, 2010 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-20570710

RESUMO

We have identified a specific LHR mRNA binding protein that selectively binds to the polypyrimidine-rich bipartite sequence in the coding region of the LHR mRNA and accelerates its degradation. This process has been shown to be one of the mechanisms that is responsible for the loss of the steady-state levels of LHR mRNA following the preovulatory LH surge or the down regulation of the receptor in response to the administration of a pharmacological dose of LH or hCG. The trans factor, designated as the LHR mRNA binding protein (LRBP), was purified and its identity was established as being mevalonate kinase, an enzyme involved in cholesterol biosynthesis. When mevalonate kinase expression was abolished by treating cultured luteal cells with 25-hydroxycholesterol, the ability to undergo LH-induced down regulation of LHR mRNA was completely abrogated. Examination of the crystal structure of mevalonate kinase coupled with mutagenesis of the critical residues in the catalytic site revealed that the catalytic site is in close proximity to the LHR mRNA binding site. Further studies revealed that mevalonate kinase causes LHR mRNA degradation by acting as a translational suppressor by forming an untranslatable ribonucleoprotein (RNP) complex which is then targeted for degradation. These studies show that LHR expression in the ovary is regulated by a post-transcriptional mechanism mediated by mevalonate kinase thereby linking LHR expression with cholesterol metabolism.


Assuntos
Regulação da Expressão Gênica/fisiologia , Receptores da Gonadotropina/genética , Esteróis/metabolismo , Humanos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores do LH/metabolismo
18.
Fertil Steril ; 93(6): 2042-7, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19342033

RESUMO

OBJECTIVE: To determine the effect of thiazolidenedione derivatives (TZDs) on vascular endothelial growth factor (VEGF) production by human luteinized granulosa cells and the morphologic development of murine embryos. DESIGN: Prospective, experimental, in vitro and in vivo study. SETTING: Research laboratory. PATIENT(S): Follicular aspirates from 10 women undergoing oocyte retrieval. INTERVENTION(S): Isolated human granulosa cells were treated with a dimethyl sulfoxide (DMSO) control or ciglitazone, in the presence and absence of an hCG stimulus. Embryos extracted from superovulated B6C3F1 female mice were cultured in the presence of DMSO or pioglitazone. MAIN OUTCOME MEASURE(S): Vascular endothelial growth factor concentrations at 24 and 48 hours. Morphologic development of murine embryos at 96 hours. RESULT(S): Following an hCG stimulus, treatment with 20 microM or 40 microM ciglitazone decreased VEGF production in a statistically significant manner at both time intervals. Blastocyst development at 96 hours did not significantly differ between untreated zygotes and those treated with pioglitazone. CONCLUSION(S): Ciglitazone significantly decreased VEGF production by human granulosa cells in an in vitro model. Pioglitazone did not adversely impact the development of cultured murine embryos. Although mechanistic evidence is not provided, the pivotal role of VEGF in ovarian hyperstimulation syndrome prompts investigation of TZDs as a novel treatment for this condition.


Assuntos
Células da Granulosa/efeitos dos fármacos , Luteinização/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Técnicas de Cultura Embrionária , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Células da Granulosa/metabolismo , Células da Granulosa/fisiologia , Humanos , Hipoglicemiantes/farmacologia , Luteinização/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo
19.
Endocrinology ; 150(2): 929-35, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18927218

RESUMO

FSH, acting through multiple signaling pathways, regulates the proliferation and growth of granulosa cells, which are critical for ovulation. The present study investigated whether AMP-activated protein kinase (AMPK), which controls the energy balance of the cell, plays a role in FSH-mediated increase in granulosa cell proliferation. Cells isolated from immature rat ovaries were grown in serum-free, phenol red free DMEM-F12 and were treated with FSH (50 ng/ml) for 0, 5, and 15 min. Western blot analysis showed a significant reduction in AMPK activation as observed by a reduction of phosphorylation at thr 172 in response to FSH treatment at all time points tested. FSH also reduced AMPK phosphorylation in a dose-dependent manner with maximum inhibition at 100 ng/ml. The chemical activator of AMPK (5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside, 0.5 mm) increased the cell cycle inhibitor p27 kip expression significantly, whereas the AMPK inhibitor (compound C, 20 microm) and FSH reduced p27kip expression significantly compared with control. FSH treatment resulted in an increase in the phosphorylation of AMPK at ser 485/491 and a reduction in thr 172 phosphorylation. Inhibition of Akt phosphorylation using Akt inhibitor VIII reversed the inhibitory effect of FSH on thr 172 phosphorylation of AMPK, whereas ERK inhibitor U0126 had no effect. These results show that FSH, through an Akt-dependent pathway, phosphorylates AMPK at ser 481/495 and inhibits its activation by reducing thr 172 phosphorylation. AMPK activation by 5-amino-imidazole-4-carboxamide-1-beta-D-ribofuranoside treatment resulted in a reduction of cell cycle regulatory protein cyclin D2 mRNA expression, whereas FSH increased the expression by 2-fold. These results suggest that FSH promotes granulosa cell proliferation by increasing cyclin D2 mRNA expression and by reducing p27 kip expression by inhibiting AMPK activation through an Akt-dependent pathway.


Assuntos
Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Proteína Oncogênica v-akt/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Técnicas de Cultura de Células , Células Cultivadas , Ciclina D2 , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Ciclinas/genética , Ciclinas/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Células da Granulosa/metabolismo , Proteína Oncogênica v-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
20.
Mol Endocrinol ; 21(9): 2233-41, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17550979

RESUMO

Posttranscriptional mechanism plays a crucial role in regulating LH receptor (LHR) expression in the ovary. We have identified a novel trans-factor, LHR mRNA binding protein (LRBP), which binds to a polypyrimidine-rich bipartite sequence of the coding region of LHR mRNA, and its identity was established as mevalonate kinase (Mvk). Although an inverse relation between LHR mRNA expression and RNA binding activity of LRBP has been established, its intermediary role in LHR mRNA expression has not been demonstrated. The present study examined the direct role of Mvk in regulating LHR expression by using primary cultures of human granulosa cells as a model system. A marked decrease in LHR mRNA stability and an increase in Mvk expression were seen when cultured granulosa cells were treated with human chorionic gonadotropin (hCG) in vitro. This treatment also resulted in an increase in LHR mRNA binding activity in the cytosolic fractions prepared from hCG-treated cells compared with the control. Because Mvk expression is regulated by sterol response element-binding protein-1, which is sensitive to the cellular concentration of 25-hydroxycholesterol (25-OHC), cultured granulosa cells were treated with this oxysterol, and the expression of Mvk gene was examined. As expected, treatment with 25-OHC inhibited the Mvk (LRBP) expression, as well as the LHR mRNA binding activity of LRBP. To determine the role of Mvk in ligand-mediated down-regulation of LHR mRNA, cells were additionally treated with 25-OHC when treated with hCG. The results showed that the decrease in Mvk expression by oxysterol treatment abrogated ligand-induced down-regulation of LHR mRNA. These results therefore establish a direct participation of Mvk in regulating LHR expression and suggest a novel relationship between cholesterol metabolism and LHR expression in the ovary.


Assuntos
Células da Granulosa/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Proteínas de Ligação a RNA/fisiologia , Receptores do LH/genética , Esteróis/metabolismo , Adulto , Células Cultivadas , Gonadotropina Coriônica/fisiologia , Feminino , Humanos , RNA Mensageiro/metabolismo , Receptores do LH/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA