Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Fertil Steril ; 103(3): 826-33, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25516084

RESUMO

OBJECTIVE: To explore the temporal expression in granulosa and theca cells of key members of the MMP and ADAMTS families across the periovulatory period in women to gain insight into their possible roles during ovulation and early luteinization. DESIGN: Experimental prospective clinical study and laboratory-based investigation. SETTING: University medical center and private IVF center. ANIMAL AND PATIENT(S): Thirty-eight premenopausal women undergoing surgery for tubal ligation and six premenopausal women undergoing assisted reproductive techniques. INTERVENTION(S): Administration of hCG and harvesting of follicles by laparoscopy and collection of granulosa-lutein cells at oocyte retrieval. MAIN OUTCOME MEASURE(S): Expression of mRNA for matrix metalloproteinase (MMPs) and the A disintegrin and metalloproteinase with thrombospondin-like motifs (ADAMTS) in human granulosa cells and theca cells collected across the periovulatory period of the menstrual cycle and in cultured granulosa-lutein cells after hCG. Localization of MMPs and ADAMTSs by immunohistochemistry. RESULT(S): Expression of MMP1 and MMP19 mRNA increased in both granulosa and theca cells after hCG administration. ADAMTS1 and ADAMTS9 mRNA increased in granulosa cells after hCG treatment, however, thecal cell expression for ADAMTS1 was unchanged, while ADAMTS9 expression was decreased. Expression of MMP8 and MMP13 mRNA was unchanged. Immunohistochemistry confirmed the localization of MMP1, MMP19, ADAMTS1, and ADAMTS9 to the granulosa and thecal cell layers. CONCLUSION(S): The collection of the dominant follicle throughout the periovulatory period has allowed the identification of proteolytic remodeling enzymes in the granulosa and theca compartments that may be critically involved in human ovulation. These proteinases may work in concert to regulate breakdown of the follicular wall and release of the oocyte.


Assuntos
Gonadotropina Coriônica/farmacologia , Fase Folicular , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Peptídeo Hidrolases/biossíntese , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Células Cultivadas , Indução Enzimática/efeitos dos fármacos , Feminino , Fase Folicular/efeitos dos fármacos , Fase Folicular/genética , Fase Folicular/metabolismo , Humanos , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Folículo Ovariano/enzimologia
2.
Biol Reprod ; 88(1): 18, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23136296

RESUMO

A local autocrine/paracrine role for progesterone is an absolute requirement for corpus luteum formation in primates. Despite this, the mechanism(s) remain obscure, although existing data suggest an anti-apoptotic action to be central. There are a limited number of progestin-regulated gene targets identified in the luteinizing primate follicle, suggesting that a small number of important genes may mediate progesterone action. Possible gene targets could be the epidermal growth factor (EGF) family members amphiregulin (AREG) and epiregulin (EREG). Using macaques undergoing controlled ovarian stimulation cycles, we show that the phosphorylation of EGF receptor (EGFR), ERK 1/2, and AKT increases 6 h after an ovulatory human chorionic gonadotropin (hCG) stimulus and remains activate through 24 h. Immunoreactive EREG and AREG ligands in the follicular fluid both increased in a time frame commensurate with EGFR phosphorylation. The mRNA expression of AREG and EREG in nonluteinized granulosa cells (NLGC) was induced in culture with hCG, an effect blocked by progesterone receptor (PGR) antagonists. Overexpression of PGR B in NLGC and treatment with a nonmetabolizable progestin did not increase either gene, indicating both progesterone and luteinizing hormone/CG are necessary. Addition of EGF and EGF-like ligands did not promote steroidogenesis in vitro by granulosa cells in the presence of gonadotropin, but were able to partially reverse RU486-induced cell death. These data suggest that progesterone promotes the expression of AREG and EREG, which in turn maintain viability of luteinizing granulosa cells, representing one possible mechanism whereby progesterone promotes corpus luteum formation in the primate.


Assuntos
Apoptose/fisiologia , Fator de Crescimento Epidérmico/fisiologia , Células da Granulosa/efeitos dos fármacos , Macaca , Progesterona/metabolismo , Animais , Gonadotropina Coriônica/farmacologia , Feminino , Líquido Folicular , Regulação da Expressão Gênica/fisiologia , Células da Granulosa/fisiologia , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcriptoma
4.
Alcohol Clin Exp Res ; 35(8): 1534-40, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21438889

RESUMO

BACKGROUND: The timing of the origins of fetal alcohol syndrome has been difficult to determine, in part because of the challenge associated with in vivo studies of the peri-implantation stage of embryonic development. Because embryonic stem cells (ESCs) are derived from blastocyst stage embryos, they are used as a model for early embryo development. METHODS: Rhesus monkey ESC lines (ORMES-6 and ORMES-7) were treated with 0, 0.01, 0.1, or 1.0% ethanol, 1.0% ethanol with estradiol, or 0.00025% acetaldehyde with or without estradiol for 4 weeks. RESULTS: Although control ESCs remained unchanged, abnormal morphology of ESCs in the ethanol and acetaldehyde treatment groups was observed before 2 weeks of treatment. Immunofluorescence staining of key pluripotency markers (TRA-1-81 and alkaline phosphatase) indicated a loss of ESC pluripotency in the 1.0% ethanol group. ORMES-7 was more sensitive to effects of ethanol than ORMES-6. CONCLUSIONS: Estradiol appeared to increase sensitivity to ethanol in the ORMES-6 and ORMES-7 cell line. The morphological changes and labeling for pluripotency, proliferation, and apoptosis demonstrated that how ethanol affects these early cells that develop in culture, their differentiation state in particular. The effects of ethanol may be mediated in part through metabolic pathways regulating acetaldehyde formation, and while potentially accentuated by estradiol in some individuals, how remains to be determined.


Assuntos
Acetaldeído/toxicidade , Diferenciação Celular/efeitos dos fármacos , Depressores do Sistema Nervoso Central/toxicidade , Células-Tronco Embrionárias/efeitos dos fármacos , Estradiol/toxicidade , Estrogênios/toxicidade , Etanol/toxicidade , Acetaldeído/metabolismo , Animais , Apoptose/efeitos dos fármacos , Blastocisto/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Depressores do Sistema Nervoso Central/metabolismo , Grupos Controle , Células-Tronco Embrionárias/patologia , Células-Tronco Embrionárias/fisiologia , Estradiol/metabolismo , Estrogênios/metabolismo , Etanol/metabolismo , Macaca mulatta
5.
Endocrinology ; 151(12): 5865-72, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20926582

RESUMO

An ovulatory stimulus induces the rapid and dramatic increase in progesterone synthesis by the primate ovarian follicle. However, little is known about the early events leading to the shift from estrogen to progesterone production. Because steroidogenesis represents an aspect of cholesterol metabolism, it was hypothesized that transcription factors regulating cholesterol balance would be among the earliest to change in response to an ovulatory stimulus. Granulosa cells were isolated from rhesus monkey follicles following controlled ovarian stimulation protocols before or up to 24 hr after an ovulatory human chorionic gonadotropin (hCG) bolus. The peroxisome proliferator-activated receptor-γ (PPARG) and the liver X receptors [nuclear receptor (NR)1H2, NR1H3] decreased within 3 hr of hCG, as did the reverse cholesterol transporters ATP-binding cassette (ABC)A1 and ABCG1. Treatment of granulosa cells isolated before an ovulatory stimulus with hCG and rosiglitizone resulted in an increase in NR1H3 and ABCG1, and decreased steroidogenic acute regulatory (STAR) protein and scavenger receptor-BI (SCARB1). A liver X receptor agonist attenuated hCG-induced progesterone synthesis in vitro and increased the expression of ABCA1 and ABCG1, and suppressed STAR, P450 side-chain cleavage A1, hydroxysteroid dehydrogenase 3B, and SCARB1. These data suggest that an initial action of LH/CG on the primate preovulatory follicle is to rapidly reduce the expression of PPARG, resulting in reduced NR1H3 with the consequence shifting the balance from cholesterol efflux via ABCA1 and ABCG1 to cholesterol uptake (SCARB1) and metabolism (STAR, P450 side-chain cleavage A1, hydroxysteroid dehydrogenase 3B). That the regulation of PPARG and the liver X receptors occurs within 3 hr strongly indicates that early events in the primate luteinizing follicle are critical to successful ovulation and luteal formation.


Assuntos
Gonadotropina Coriônica/farmacologia , Regulação para Baixo/efeitos dos fármacos , Ovulação/fisiologia , PPAR gama/metabolismo , Progesterona/biossíntese , Animais , Colesterol/metabolismo , Feminino , Perfilação da Expressão Gênica , Células da Granulosa , Humanos , Receptores X do Fígado , Macaca , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , PPAR gama/genética , Rosiglitazona , Tiazolidinedionas/farmacologia
6.
Endocrinology ; 151(11): 5519-27, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20844007

RESUMO

Our goal was to examine the effect of deficiency of the lipoprotein receptor, scavenger receptor class B type I (SR-BI), on progesterone secretion in human granulosa cells (HGL5). Scrambled or SR-BI small interfering RNA [knockdown (KD)] cells were exposed to dimethylsulfoxide [DMSO, vehicle for forskolin (Fo)], Fo, serum, high-density lipoprotein, low-density lipoprotein (LDL), or Fo plus lipoproteins or serum for 24 h. Progesterone secretion was lower in all of the SR-BI KD cells regardless of treatment. We examined progesterone secretion in SR-BI KD, LDL receptor KD, and double KD cells incubated with DMSO, Fo, LDL, or Fo + LDL for 6-24 h. As compared with scrambled cells, progesterone secretion was lower in SR-BI and double KD cells regardless of treatment; whereas progesterone secretion was only lower in LDL receptor KD cells incubated with LDL and Fo + LDL. We measured phosphorylation of hormone-sensitive lipase (pHSL) expression, intracellular total cholesterol (TC) mass, and progesterone secretion in scrambled and SR-BI KD cells incubated with DMSO or Fo for 2-24 h. The expression of pHSL was similar between the cells and conditions. The mean change in TC mass and progesterone secretion was lower in SR-BI KD cells exposed to DMSO and Fo. Incubating SR-BI KD cells with 22-hydroxy cholesterol did not overcome the reduction in progesterone secretion. At different time points, RNA expression of steroidogenic acute regulatory protein, side-chain cleavage, and 3ß-hydroxysteroid dehydrogenase was significantly lower in SR-BI KD cells incubated with Fo. In conclusion, SR-BI protein deficiency, in part, might explain progesterone deficiency in some infertile women.


Assuntos
Células da Granulosa/metabolismo , Progesterona/metabolismo , Receptores Depuradores Classe B/metabolismo , Western Blotting , Linhagem Celular , Células Cultivadas , Feminino , Células da Granulosa/efeitos dos fármacos , Humanos , Lipoproteínas HDL/metabolismo , Lipoproteínas HDL/farmacologia , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , RNA Interferente Pequeno , Radioimunoensaio , Receptores de LDL/genética , Receptores de LDL/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Depuradores Classe B/genética
7.
Am J Physiol Endocrinol Metab ; 296(6): E1392-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19293332

RESUMO

During hormonally induced ovarian follicle growth, granulosa cell proliferation increases and returns to baseline prior to the administration of an ovulatory stimulus. Several key genes appear to follow a similar pattern, including the luteinizing hormone receptor (LHCGR), suggesting an association between cell cycle progression and gene expression. The expression of LHCGR mRNA in granulosa cells isolated from immature rats and treated in culture with FSH increased in a time-dependent manner, whereas administration of the cell cycle inhibitor mimosine completely suppressed expression. Although forskolin was able to induce luteinization in cells treated with mimosine, human chorionic gonadotropin had no effect, indicating the functional loss of LHCGR. The effects of mimosine on cell cycle progression and LHCGR mRNA expression were reversible within 24 h of mimosine removal. Cell cycle inhibition did not alter the stability of LHCGR mRNA, indicating that the primary effect was at the transcriptional level. To determine whether the relationship between LHCGR expression and cell cycle were relevant in vivo, immature rats were given a bolus of PMSG, followed by a second injection of either saline or PMSG 24 h later to augment levels of proliferation. The expression of LHCGR mRNA was elevated in the ovaries of animals receiving a supplement of PMSG. Mimosine also blocked cell cycle progression and LHCGR mRNA expression in macaque granulosa cells isolated following controlled ovarian stimulation cycles and in two different mouse Leydig tumor lines. These data collectively indicate that LHCGR mRNA is expressed as a function of the passage of cells across the G1-S phase boundary.


Assuntos
Fase G1/fisiologia , Células da Granulosa/citologia , Células da Granulosa/fisiologia , Receptores do LH/genética , Fase S/fisiologia , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Gonadotropina Coriônica/farmacologia , Colforsina/farmacologia , Feminino , Fase G1/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Gonadotropinas Equinas/farmacologia , Células da Granulosa/efeitos dos fármacos , Tumor de Células de Leydig , Macaca mulatta , Masculino , Camundongos , Mimosina/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fase S/efeitos dos fármacos , Neoplasias Testiculares
8.
Am J Physiol Endocrinol Metab ; 296(5): E1049-58, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19276391

RESUMO

The developmental competence of in vitro-matured (IVM) rhesus macaque cumulus oocyte complexes (COCs) is deficient compared with in vivo-matured (IVM) oocytes. To improve oocyte quality and subsequent embryo development following IVM, culture conditions must be optimized. A series of experiments was undertaken to determine the role of epidermal growth factor (EGF) during IVM of rhesus macaque COCs. The addition of Tyrphostin AG-1478 (a selective inhibitor of the EGF receptor EGFR) to the IVM medium yielded fewer oocytes maturing to metaphase II of meiosis II (MII), decreased cumulus expansion, and a lower percentage of embryos that developed to the blastocyst stage compared with untreated IVM controls, indicating that EGFR activation is important for IVM maturation in the rhesus macaque. However, the addition of recombinant human EGF (r-hEGF) to the IVM medium did not enhance outcome. The expression of mRNAs encoding the EGF-like factors amphiregulin, epiregulin, and betacellulin in cumulus cells indicates that these factors produced by cumulus cells may be responsible for maximal EGFR activation during oocyte maturation, precluding any further effect of exogenous r-hEGF. Additionally, these results illustrate the potential futility of exogenous supplementation of IVM medium without prior knowledge of pathway activity.


Assuntos
Células do Cúmulo/fisiologia , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/metabolismo , Macaca mulatta/fisiologia , Oócitos/fisiologia , Anfirregulina , Animais , Betacelulina , Células do Cúmulo/efeitos dos fármacos , Família de Proteínas EGF , Desenvolvimento Embrionário/fisiologia , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/biossíntese , Fator de Crescimento Epidérmico/genética , Epirregulina , Receptores ErbB/antagonistas & inibidores , Feminino , Fertilização in vitro/veterinária , Glicoproteínas/biossíntese , Glicoproteínas/genética , Imuno-Histoquímica/veterinária , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Oócitos/citologia , Oócitos/efeitos dos fármacos , Fosforilação , Gravidez , Quinazolinas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Transdução de Sinais , Tirfostinas/farmacologia
9.
Endocrinology ; 150(2): 957-65, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18832102

RESUMO

An ovulatory hCG stimulus to rhesus macaques undergoing controlled ovarian stimulation protocols results in a rapid and sustained increase in progesterone synthesis. The use of lipoproteins as a substrate for progesterone synthesis remains unclear, and the expression of lipoprotein receptors [very-low-density lipoprotein receptor (VLDLR), low-density lipoprotein receptor (LDLR), and scavenger receptor-BI (SR-BI)] soon after human chorionic gonadotropin (hCG) (<12 h) has not been characterized. This study investigated lipoprotein receptor expression and lipoprotein (VLDL, LDL, and HDL) support of steroidogenesis during luteinization of macaque granulosa cells. Granulosa cells were aspirated from rhesus monkeys undergoing controlled ovarian stimulation before or up to 24 h after an ovulatory hCG stimulus. The expression of VLDLR decreased within 3 h of hCG, whereas LDLR and SR-BI increased at 3 and 12 h, respectively. Granulosa cells isolated before hCG were cultured for 24 h in the presence of FSH or FSH plus hCG with or without VLDL, LDL, or HDL. Progesterone levels increased in the presence of hCG regardless of lipoprotein addition, although LDL, but not HDL, further augmented hCG-induced progesterone. Other cells were cultured with FSH or FSH plus hCG without an exogenous source of lipoprotein for 24 h, followed by an additional 24 h culture with or without lipoproteins. Cells treated with hCG in the absence of any lipoprotein were unable to maintain progesterone levels through 48 h, whereas LDL (but not HDL) sustained progesterone synthesis. These data suggest that an ovulatory stimulus rapidly mobilizes stored cholesterol esters for use as a progesterone substrate and that as these are depleted, new cholesterol esters are obtained through an LDLR- and/or SR-BI-mediated mechanism.


Assuntos
Antígenos CD36/genética , Células da Granulosa/efeitos dos fármacos , Lipoproteínas/farmacologia , Luteinização/efeitos dos fármacos , Receptores de LDL/genética , Esteroides/biossíntese , Animais , Antígenos CD36/metabolismo , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Estrogênios/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/metabolismo , Luteinização/genética , Luteinização/metabolismo , Macaca mulatta , Progesterona/metabolismo , Receptores de LDL/metabolismo
10.
Mol Cell Endocrinol ; 233(1-2): 15-24, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15767042

RESUMO

PPARgamma is expressed in both the rodent and human ovary, but the endogenous activation state of PPARgamma in the ovary and its normal role in ovarian function remain unclear. Here, we investigated mRNA and protein expression as well as DNA-binding activity of PPARgamma during follicle growth and luteinization in the immature, gonadotropin-primed rat model. Gel shift analysis demonstrated binding of ovarian PPAR to a consensus peroxisome proliferator response element (PPRE) that was supershifted with an antibody specific for PPARgamma, but not with antibodies specific for PPARalpha or beta/delta. PPARgamma expression and DNA-binding activity was highest 0-12 h post-PMSG, but declined during later stages of follicle growth (24-36 h post-PMSG). Administration of hCG induced a decline in PPARgamma mRNA, protein, and DNA-binding activity beginning at 4 h. Treatment of preovulatory granulosa cells with the PPARgamma ligand troglitazone (1-10 microM) in vitro decreased cell viability, increased sub-G1 apoptosis, and reduced DNA synthesis. Troglitazone induced p53 protein expression and decreased bcl-2 mRNA, suggesting possible mechanisms for troglitazone-induced apoptosis. These data indicate that PPARgamma is in the ovary is capable of binding DNA in the absence of pharmacological activation and provide evidence for a possible physiologic role for this receptor in regulating granulosa cell survival.


Assuntos
Apoptose , Cromanos/farmacologia , Proteínas de Ligação a DNA/metabolismo , Células da Granulosa/efeitos dos fármacos , Folículo Ovariano/crescimento & desenvolvimento , PPAR gama/metabolismo , Tiazolidinedionas/farmacologia , Animais , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Ligantes , Folículo Ovariano/citologia , PPAR gama/agonistas , PPAR gama/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Ratos , Elementos de Resposta , Troglitazona , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
11.
Endocrinology ; 145(12): 5734-44, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15331571

RESUMO

Ovulation and formation of a functional corpus luteum in primates involve cascades of events, including increased progesterone synthesis and changes in granulosa cell proliferation. However, critical gaps remain in our understanding of how an ovulatory gonadotropin surge initiates these processes. To more fully elucidate changes in the cell cycle during luteal formation, the actions of the tumor suppressor p53 were examined. Rhesus macaque granulosa cells were isolated during controlled ovarian stimulation protocols before (nonluteinized) or after (luteinized) an ovulatory gonadotropin stimulus. Phosphorylated p53 protein was detected in the cytoplasm of granulosa cells before and after human chorionic gonadotropin (hCG) treatment, whereas granulosa cells from hormonally controlled rats did not express p53 before or after hCG. Treatment of nonluteinized macaque granulosa cells with hCG and the p53 inhibitor pifithrin-alpha (PFT) in vitro did not alter markers of the cell cycle, including proliferating cell nuclear antigen, p21, and human double minute (HDM)-2 expression compared with hCG alone. Levels of pregnenolone and progesterone increased 2- and 4-fold, respectively, within 6 h of hCG treatment, whereas PFT completely blocked this hCG-induced effect. Estradiol was increased transiently (>10-fold) by hCG plus PFT relative to levels after hCG alone. PFT also inhibited hCG-induced increases in steroidogenic acute regulatory protein and 3beta-hydroxysteroid dehydrogenase mRNAs. Similar results were obtained using the human adrenocortical cell line H295R, suggesting that p53 may have a general function in primate steroidogenesis. These data indicate that p53 plays a key role in luteinization of the primate ovarian follicle though the regulation of steroidogenic enzymes leading to progesterone synthesis.


Assuntos
Córtex Suprarrenal/fisiologia , Células da Granulosa/fisiologia , Progesterona/biossíntese , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Córtex Suprarrenal/citologia , Animais , Divisão Celular/fisiologia , Células Cultivadas , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Feminino , Expressão Gênica , Células da Granulosa/citologia , Humanos , Macaca mulatta , Ovulação/fisiologia , Fosfoproteínas/genética , RNA Mensageiro/análise
12.
Biol Reprod ; 71(1): 366-73, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-14985242

RESUMO

During the periovulatory interval, intrafollicular progesterone (P) prevents follicular atresia and promotes ovulation. Whether P influences oocyte quality or maturation and follicle rupture independent of the midcycle gonadotropin surge was examined. Rhesus monkeys underwent controlled ovarian stimulation with recombinant human gonadotropins followed by a) experiment 1: an ovulatory bolus of hCG alone or with a steroid synthesis inhibitor (trilostane, TRL), or TRL + the progestin R5020; or b) no hCG, but rather sesame oil (vehicle), R5020, or dihydrotestosterone (DHT). In experiment 1, the majority of oocytes remained immature (65% +/- 20%) by 12 h post-hCG. However, the percentage of degenerating oocytes increased (P < 0.05) with TRL (42% +/- 22% vs. 0% controls), but was reduced (P < 0.05) by progestin replacement (15% +/- 7%). By 36 h post-hCG, the majority of oocytes in all three groups reached metaphase II (MI). In experiment 2, no evidence of follicle rupture was observed in the vehicle, R5020, or DHT groups. Despite the absence of hCG, a significant (P < 0.05) percentage of oocytes resumed meiosis to metaphase I in R5020- (41 +/- 9) and DHT- (36 +/- 15) but not vehicle- (4 +/- 4) treated animals. Only oocytes from R5020-treated animals continued meiosis in vivo to MII. More (P < 0.05) oocytes fertilized in vitro with R5020 (40%) than with vehicle (20%) or DHT (22%). Thus, P is unable to elicit ovulation in the absence of an ovulatory gonadotropin surge; however, P and/or androgens may prevent oocyte atresia and promote oocyte nuclear maturation in primate follicles.


Assuntos
Gonadotropinas/metabolismo , Oogênese/efeitos dos fármacos , Progesterona/farmacologia , Animais , Gonadotropina Coriônica/farmacologia , Di-Hidrotestosterona/farmacologia , Fertilização in vitro , Macaca mulatta , Meiose/efeitos dos fármacos , Oócitos/citologia , Ovulação/efeitos dos fármacos , Indução da Ovulação , Progestinas/farmacologia , Promegestona/farmacologia
13.
Hum Reprod ; 18(11): 2257-63, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14585870

RESUMO

BACKGROUND: A method was sought to control ovulation of the dominant follicle and to test the importance of LH during the late follicular phase of the menstrual cycle. Menstrual cycles of rhesus monkeys were monitored, and treatment initiated at the late follicular phase (after dominant follicle selection, before ovulation). METHODS: The 2-day treatment consisted of GnRH antagonist plus either r-hFSH and r-hLH (1:1 or 2:1 dose ratio) or r-hFSH alone. In addition, half of the females received an ovulatory bolus of hCG. RESULTS: When treatment was initiated at estradiol levels >120 pg/ml, neither the endogenous LH surge, ovulation nor luteal function were controlled. However, when treatment was initiated at estradiol levels 80-120 pg/ml using either 1:1 or 2:1 dose ratios of FSH:LH, the LH surge was prevented, and ovulation occurred following hCG treatment. FSH-only treatment also prevented the LH surge, but follicle development appeared abnormal, and hCG failed to stimulate ovulation. CONCLUSIONS: Control over the naturally dominant follicle is possible during the late follicular phase using an abbreviated GnRH antagonist, FSH+LH protocol. This method offers a model to investigate periovulatory events and their regulation by gonadotrophins/local factors during the natural menstrual cycle in primates.


Assuntos
Fase Folicular/fisiologia , Hormônio Luteinizante/fisiologia , Folículo Ovariano/fisiologia , Indução da Ovulação/métodos , Animais , Gonadotropina Coriônica/farmacologia , Relação Dose-Resposta a Droga , Feminino , Hormônio Foliculoestimulante/farmacologia , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Humanos , Hormônio Luteinizante/administração & dosagem , Hormônio Luteinizante/farmacologia , Macaca mulatta , Folículo Ovariano/efeitos dos fármacos , Proteínas Recombinantes/farmacologia
14.
Endocrinology ; 144(4): 1249-56, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12639907

RESUMO

Granulosa cell luteinization involves the attenuation of gonadotropin-induced proliferation. Although recent evidence indicates that primate granulosa cells stop dividing within 12 h of an ovulatory stimulus, early events in cell cycle arrest remain unknown. In the current study an in vitro model of primate granulosa cell luteinization is established that allows assessment of early events in terminal differentiation. A luteinizing dose of human chorionic gonadotropin (hCG) results in a secondary rise in proliferation before cell cycle arrest that is paralleled by a transient increase in the expression of c-Myc. In contrast, the c-Myc antagonists Mad1, Mad4, and Mxi1 are transiently repressed by hCG. Max, the common dimerization partner for Myc and Mad, is similarly repressed by hCG, suggesting that changes in the expression of this gene may further regulate the activity of Myc and Mad. To determine whether other cell cycle regulatory families are involved in luteinization, the expression of p53 and the wild-type p53-inducible phosphatase (wip1) was examined. Similar to Mad and Max, p53 and wip1 are transiently repressed by hCG, suggesting that the p53 and Mad pathways have either parallel or cooperative roles in luteinization. Thus, luteinization of primate granulosa cells is preceded by a burst of proliferation that is regulated by changes in the relative levels of c-Myc, Max, and Mad as well as p53.


Assuntos
Corpo Lúteo/fisiologia , Luteinização/fisiologia , Ovulação/fisiologia , Proteínas Proto-Oncogênicas c-myc/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Fatores de Transcrição de Zíper de Leucina Básica , Divisão Celular/fisiologia , Corpo Lúteo/citologia , Ciclinas/genética , Proteínas de Ligação a DNA/genética , Feminino , Expressão Gênica/fisiologia , Técnicas In Vitro , Macaca mulatta , Proteínas de Neoplasias/genética , Fosfoproteínas Fosfatases/genética , Antígeno Nuclear de Célula em Proliferação/genética , Proteína Fosfatase 2C , RNA Mensageiro/análise , Proteínas Repressoras/genética , Timidina Quinase/genética , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA