Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Reprod Toxicol ; 104: 114-124, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34311058

RESUMO

Disruption of granulosa cells (GCs), the main functional cells in the ovary, is associated with impaired female fertility. Epidemiological studies demonstrated that women have detectable levels of organic pollutants (e.g., perfluorooctanoate, perfluorooctane sulfonate, 2,2-dichlorodiphenyldichloroethylene, polychlorinated biphenyl 153, and hexachlorobenzene) in their follicular fluid (FF), and thus these compounds may directly affect the function of GCs in the ovary. Considering that humans are exposed to multiple pollutants simultaneously, we elucidated the effects of a mixture of endocrine-disrupting chemicals (EDCs) on human granulosa HGrC1 cells. The EDC mixture directly increased progesterone secretion by upregulating 3ß-hydroxysteroid dehydrogenase (3ßHSD) expression. Furthermore, the EDC mixture increased activity of mitochondria, which are the central sites for steroid hormone biosynthesis, and the ATP content. Unexpectedly, the EDC mixture reduced glucose transporter 4 (GLUT4) expression and perturbed glucose uptake; however, this did not affect the glycolytic rate. Moreover, inhibition of GLUT1 by STF-31 did not alter the effects of the EDC mixture on steroid secretion but decreased basal estradiol secretion. Taken together, our results demonstrate that the mixture of EDCs present in FF can alter the functions of human GCs by disrupting steroidogenesis and may thus adversely affect female reproductive health. This study highlights that the EDC mixture elicits its effects by targeting mitochondria and increases mitochondrial network formation, mitochondrial activity, and expression of 3ßHSD, which is associated with the inner mitochondrial membrane.


Assuntos
Líquido Folicular/metabolismo , Poluentes Orgânicos Persistentes/metabolismo , Progesterona/metabolismo , Disruptores Endócrinos/metabolismo , Estradiol/metabolismo , Feminino , Líquido Folicular/química , Células da Granulosa/efeitos dos fármacos , Humanos , Luteinização/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Neoplasias Ovarianas , Poluentes Orgânicos Persistentes/toxicidade , Esteroides/metabolismo , Regulação para Cima/efeitos dos fármacos
2.
Mol Cell Endocrinol ; 520: 111080, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33189865

RESUMO

During obesity, excess body weight is not only associated with an increased risk of type 2-diabetes, but also several other pathological processes, such as infertility. Adipose tissue is the largest endocrine organ of the body that produces adipokines, including adiponectin. Adiponectin has been reported to control fertility through the hypothalamic-pituitary-gonadal axis, and folliculogenesis in the ovaries. In this study, we focused on a recent adiponectin-like synthetic agonist called AdipoRon, and its action in human luteinized granulosa cells. We demonstrated that AdipoRon activated the adenosine monophosphate-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor alpha (PPAR) signalling pathways in human luteinized granulosa cells. A 25 µM AdipoRon stimulation reduced granulosa cell proliferation by inducing cell cycle arrest in G1, associated with PTEN and p53 pathway activation. In addition, AdipoRon perturbed cell metabolism by decreasing mitochondrial activity and ATP production. In human luteinized granulosa cells, AdipoRon increased phosphodiesterase activity, leading to a drop in cyclic adenosine monophosphate (cAMP) production, aromatase expression and oestrogens secretion. In conclusion, AdipoRon impacted folliculogenesis by altering human luteinized granulosa cell function, via steroid production and cell proliferation. This agonist may have applications for improving ovarian function in metabolic disorders or granulosa cancers.


Assuntos
Adiponectina/agonistas , Células da Granulosa/metabolismo , Luteinização/metabolismo , Piperidinas/farmacologia , Esteroides/biossíntese , Proteínas Quinases Ativadas por AMP/metabolismo , Adiponectina/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Feminino , Células da Granulosa/efeitos dos fármacos , Humanos , Luteinização/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , PTEN Fosfo-Hidrolase/metabolismo , Receptores Ativados por Proliferador de Peroxissomo , Piperidinas/química , Proteínas Proto-Oncogênicas c-akt/metabolismo
3.
PLoS One ; 15(9): e0238814, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32881970

RESUMO

Prostate cancer (PCa) cells become castrate-resistant after initial tumor regression following castration-based lowering of testosterone (T). De-novo intra-tumoral steroid synthesis is a suggested biological mechanism of castration resistant PCa, but the regulators are unknown. Testicular T production is controlled by the luteinizing hormone/choriogonadotropin receptor (LHCGR). To elucidate the influence of LHCGR on PCa development the presence and effects of LHCGR in PCa and whether LHCGR in serum holds prognostic information in PCa patients is investigated. LHCGR expression was investigated by RT-PCR, WB, IHC, qPCR in PCa cell lines and prostatic tissue. Steroid production was measured in media from cell lines with LC-MS/MS and expression of steroidogenic enzymes with qPCR. Serum LHCGR (sLHCGR) was measured with ELISA in PCa patients (N = 157). Presence of LHCGR was established in prostatic tissue and PCa cell lines. Cell proliferation increased by 1.29-fold in LNCaP (P = 0.007) and 1.33-fold in PC-3 cells (P = 0.0007), when stimulated by luteinizing hormone. Choriogonadotropin stimulation decreased proliferation 0.93-fold in DU145 cells (P = 0.05), but none of the treatments altered steroid metabolite secretion. Low sLHCGR concentration was associated with a higher risk of biochemical failure after radical prostatectomy (HR = 3.05, P = 0.06) and castration resistance (HR = 6.92, P = 0.004) compared to high sLHCGR concentration. LHCGR is expressed in PCa and may exert a growth regulatory role in PCa derived cell lines. A potential prognostic role of sLHCGR for determining recurrence risk in PCa patients is found in this pilot study but needs verification in larger cohorts.


Assuntos
Hormônio Luteinizante/metabolismo , Neoplasias da Próstata/fisiopatologia , Receptores do LH/metabolismo , Idoso , Gonadotropina Coriônica/farmacologia , Humanos , Luteinização/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Próstata/patologia , Prostatectomia/efeitos adversos , Testículo/metabolismo , Testosterona/metabolismo , Células Tumorais Cultivadas
4.
Fertil Steril ; 111(4): 641-649, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30929721

RESUMO

The advent of third party parenting ushered in the era of artificial stimulation of the endometrium. Initially intended only for patients with ovarian failure, exogenous induction of endometrial receptivity was quickly shown to be as good as natural endometrial preparation, with the advantage that the timing of embryo transfer could be controlled. It is perhaps surprising that even though the ovary produces a variety of steroids, that estradiol (E2) and progesterone (P) alone would be needed to achieve optimal receptivity; no other substance has ever been shown to improve on the basic regimen of E2 and P. A variety of routes of administration are available for both E2 and P and physiologic (or supraphysiologic) serum or endometrial tissue levels of both can be achieved. The optimal duration of E2 stimulation and the timing of the onset of P administration continue to be debated, but it appears that imitating the sequence that normally occurs in nature leads to optimal results. The poorly responsive endometrium and cases of recurrent implantation failure remain a challenge, but the clear majority of patients can successfully achieve pregnancy as long as embryos of adequate quality are transferred.


Assuntos
Implantação do Embrião/fisiologia , Transferência Embrionária/métodos , Endométrio , Luteinização , Técnicas de Reprodução Assistida , Mães Substitutas , Criopreservação , Implantação do Embrião/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Endométrio/fisiologia , Estradiol/farmacologia , Estradiol/uso terapêutico , Feminino , Fármacos para a Fertilidade Feminina/farmacologia , Fármacos para a Fertilidade Feminina/uso terapêutico , Humanos , Luteinização/efeitos dos fármacos , Luteinização/fisiologia , Gravidez , Taxa de Gravidez
5.
Reprod Fertil Dev ; 31(4): 698-704, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30414623

RESUMO

High mobility group AT-hook 1 (HMGA1) is able to regulate gene expression and function as a tumour suppressor. The spatiotemporal expression pattern of HMGA1 was investigated in this study. Immature female rats (22-23 days old) were treated with 10IU, s.c., pregnant mare's serum gonadotrophin to stimulate follicular development, followed 48h later by injection with 5IU, s.c., human chorionic gonadotrophin (hCG). Whole ovaries or granulosa cells were collected at various times after hCG administration (n=3 per time point). Real-time polymerase chain reaction and western blot analysis revealed that HMGA1 was highly stimulated in the ovary by 4-12h after hCG treatment. In situ hybridisation analysis demonstrated that Hmga1 mRNA expression was induced in granulosa cells between 8 and 12h after hCG treatment. There was negligible Hmga1 mRNA signal observed in newly forming corpora lutea. In addition, the data indicated that both the protein kinase (PK) A and PKC pathways regulated Hmga1 expression in rat granulosa cells. In rat granulosa cell cultures, upregulation of Hmga1 was dependent on new protein synthesis because Hmga1 was inhibited by cycloheximide. Furthermore, Hmga1 mRNA expression in rat granulosa cell cultures was inhibited by AG1478, whereas NS398 and RU486 had no effect, suggesting that Hmga1 expression was regulated, in part, by the epidermal growth factor pathway. In summary, the findings of this study suggest that induction of Hmga1 may be important for theca and granulosa cell differentiation into luteal cells.


Assuntos
Diferenciação Celular/fisiologia , Proteína HMGA1a/metabolismo , Luteinização/metabolismo , Ovário/metabolismo , Ovulação/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/metabolismo , Feminino , Regulação da Expressão Gênica , Gonadotropinas Equinas/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Proteína HMGA1a/genética , Luteinização/efeitos dos fármacos , Luteinização/genética , Ovulação/efeitos dos fármacos , Ovulação/genética , Ratos , Ratos Sprague-Dawley , Células Tecais/efeitos dos fármacos , Células Tecais/metabolismo
6.
Reprod Biol ; 18(1): 1-4, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29317175

RESUMO

Premature rise of progesterone during the late follicular phase in stimulated IVF cycles is a frequent event and its effect on the endometrial receptivity and on the ART (Assisted Reproductive Technique) - outcome has become a matter of intense debate and research. An emerging body of evidence demonstrates that premature progesterone rise does have a negative impact on the outcome of the ART-success. Until now, the exact cause of progesterone elevation is not fully clear, however lately published studies points to the fact, that premature progesterone elevation might be caused by enhanced FSH stimulation. The impact of elevated peripheral progesterone levels seems to be mainly on the endometrium and the window of implantation, leading to an asynchrony between the endometrium and the developing embryo. Hence, new data show additional an influence on the embryo quality. This review aims to summarize the up-to-date knowledge on the causes of premature progesterone rise during hormonal stimulation, on its influence on endometrial receptivity and embryo quality, on the impact on pregnancy and live birth rates as well as on the possible strategies to prevent this event or to deal with premature progesterone elevation in case it could not be avoided.


Assuntos
Transferência Embrionária , Fármacos para a Fertilidade Feminina/farmacologia , Infertilidade Feminina/terapia , Indução da Ovulação , Progesterona/sangue , Técnicas de Reprodução Assistida , Regulação para Cima/efeitos dos fármacos , Animais , Coeficiente de Natalidade , Relação Dose-Resposta a Droga , Resistência a Medicamentos , Transferência Embrionária/efeitos adversos , Endométrio/efeitos dos fármacos , Endométrio/fisiopatologia , Feminino , Fármacos para a Fertilidade Feminina/administração & dosagem , Fármacos para a Fertilidade Feminina/efeitos adversos , Humanos , Infertilidade Feminina/fisiopatologia , Luteinização/sangue , Luteinização/efeitos dos fármacos , Indução da Ovulação/efeitos adversos , Guias de Prática Clínica como Assunto , Gravidez , Técnicas de Reprodução Assistida/efeitos adversos
7.
J Reprod Dev ; 64(2): 129-134, 2018 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-29249775

RESUMO

Although the expression of gonadotropin-releasing hormone (GnRH) in the ovaries is well established, its physiological role remains unknown. The aim of this study was to determine whether ovarian GnRH mediates the actions of human chorionic gonadotropin (hCG) in the granulosa cells of immature female rats. Follicular growth was induced by administration of pregnant mare serum gonadotropin (PMSG, 15 IU/0.15 ml) on day 25 after birth, and hCG (20 IU/0.2 ml) was administered on day 27 revealing the increase of plasma progesterone level. Primary cultures of granulosa cells were established from large follicles 2 days after PMSG treatment. Progesterone synthesis was augmented by hCG in a dose-dependent manner. Annexin A5 (ANXA5), a biomarker of GnRH, was expressed in the granulosa-luteal cells after hCG treatment, as shown by immunohistochemistry, suggesting that hCG treatment induced GnRH action. The GnRH mRNA level was increased by hCG, and treatment with GnRH agonist (GnRHa) increased ANXA5 mRNA levels in the primary cultures of granulosa cells. Concomitant incubation of GnRH (10-7 M) or GnRHa (fertirelin acetate, 10-8 M) with hCG suppressed progesterone synthesis during a 3 h incubation period. The mRNA expression of luteinizing hormone receptor (LHR) and follicle-stimulating hormone receptor (FSHR) was synergistically stimulated and suppressed by hCG and GnRHa, respectively. GnRHa stimulated p21 expression, and GnRHa and hCG synergistically reduced the mRNA expression levels of p27 and FOXO1. These data suggest that GnRH induced by LH may have a role for the LH-mediated luteinization of granulosa cells. In addition, ANXA5 may be involved in GnRH action. GnRH-ANXA5 would be an important mechanism in cell differentiation.


Assuntos
Gonadotropina Coriônica/farmacologia , Fármacos para a Fertilidade Feminina/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/agonistas , Células da Granulosa/efeitos dos fármacos , Luteinização/efeitos dos fármacos , Ovário/efeitos dos fármacos , Animais , Anexina A5/agonistas , Anexina A5/genética , Anexina A5/metabolismo , Biomarcadores/sangue , Biomarcadores/metabolismo , Células Cultivadas , Feminino , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Gonadotropinas Equinas/farmacologia , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Humanos , Imuno-Histoquímica , Ovário/citologia , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Progesterona/agonistas , Progesterona/antagonistas & inibidores , Progesterona/biossíntese , Progesterona/sangue , Ratos Wistar , Receptores do FSH/agonistas , Receptores do FSH/antagonistas & inibidores , Receptores do FSH/genética , Receptores do FSH/metabolismo , Receptores do LH/agonistas , Receptores do LH/genética , Receptores do LH/metabolismo
8.
Gynecol Endocrinol ; 34(3): 189-191, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28942696

RESUMO

The use of GnRH antagonists (GnRHant) is increasing in the ovarian stimulation protocol. Among several other benefits, GnRHant should prevent a premature luteinization and premature ovulation, the first described either as a 'reassuringly rare event' or 'frequent event', while the second as occurring more frequently in women with decreased ovarian reserve, advanced age and poor ovarian response. Two cases of associated premature luteinization and premature ovulation, during treatment with gonadotropins and GnRHant in IVF cycles, are here reported. In both cases, premature luteinization occurred and ovulation took place during ovarian stimulation protocols with exogenous gonadotropins and GnRHant, before reaching the criteria of hCG administration, regardless of the age of the patients and their ovarian reserve. Ovulation was documented by the disappearance of most of the developing follicles, by the transformation of endometrium from a triple line picture into a uniform hyper-echogenic image, by the presence of fluid in the pouch of Douglas, by the increase of progesterone plasma levels and the simultaneous reduction of estradiol plasma levels. This evidence can be important for a correct counseling with infertile patients in preparation for an IVF cycle.


Assuntos
Fertilização in vitro/métodos , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Gonadotropinas/administração & dosagem , Antagonistas de Hormônios/administração & dosagem , Luteinização/efeitos dos fármacos , Indução da Ovulação/métodos , Ovulação/efeitos dos fármacos , Adulto , Endométrio/diagnóstico por imagem , Feminino , Humanos , Pessoa de Meia-Idade , Folículo Ovariano/diagnóstico por imagem , Ultrassonografia
9.
Hum Reprod ; 32(3): 643-652, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28158500

RESUMO

STUDY QUESTION: Can granulosa cells produce progesterone (P) in response to FSH stimulation? SUMMARY ANSWER: FSH actively promotes P synthesis and output from granulosa cells without luteinization by up-regulating the expression and increasing enzymatic activity of 3ß-hydroxysteriod dehydrogenoase (3ß-HSD), which converts pregnenolone to P. WHAT IS KNOWN ALREADY: Serum P level may rise prematurely prior to ovulation trigger in stimulated IVF cycles and adversely affect implantation and clinical pregnancy rates by impairing endometrial receptivity. STUDY DESIGN, SIZE, DURATION: A translational research study. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human ovarian cortical samples (n = 15) and non-luteinizing FSH-responsive human mitotic granulosa cell line (HGrC1) were stimulated with rec-FSH at 12.5, 25 and 50 mIU/ml concentrations for 24 and 48 h. FSH receptor expression was knocked-down and up-regulated in the granulosa cells using short hairpin RNA (shRNA) technology and activin-A administration, respectively. The expressions of the steroidogenic enzymes were analyzed at mRNA level by real-time quantitative RT-PCR, and protein level by western blot and immunoprecipitation assay. The enzymatic activity of 3ß-HSD was measured using a spectrophotometric method. In vitro estradiol (E2) and P productions of the cells before and after FSH stimulation were measured by electro-chemiluminescence immunoassay method. MAIN RESULTS AND THE ROLE OF CHANCE: Stimulation of the HGrC1 cells with FSH resulted in a dose-dependent increase in the mRNA and protein level of 3ß-HSD. Overall, when all time points and FSH doses were analyzed collectively, FSH significantly up-regulated the mRNA expression of its own receptor (3.73 ± 0.06-fold, P < 0.001), steroidogenic acute regulatory protein (stAR, 1.7 ± 0.03-fold, P < 0.01), side-chain cleavage enzyme (SCC, 1.75 ± 0.03-fold, P < 0.01), aromatase (4.49 ± 0.08-fold, P < 0.001), 3ß-HSD (1.68 ± 0.02-fold, P < 0.01) and 17ß-hydroxy steroid dehydrogenase (17ß-HSD, 2.16 ± 0.02-fold, P < 0.01) in the granulosa cells. Expression of 17α-hydroxylase (17α-OH, 1.03 ± 0.01-fold P > 0.05) did not significantly change. Similar changes were observed in the protein expression analysis of these enzymes on western blotting after FSH stimulation. FSH significantly increased 3ß-HSD, 17ß-HSD and aromatase in a dose-dependent manner but did not affect 17α-OH. Protein expression of P was increased along with 3ß-HSD after FSH stimulation, which was further evidenced by immunoprecipitation assay. Enzymatic activity of 3ß-HSD was significantly enhanced by FSH administration in the HGrC1 cells in a dose-dependent manner. In line with these findings P output (1.05 ± 0.3 vs. 0.2 ± 0.1 ng/ml, respectively, P < 0.001) from the samples stimulated with FSH were significantly increased along with E2 (1918 ± 203 vs. 932 ± 102 pg/ml, respectively, P < 0.001) compared to unstimulated controls. FSH-induced increase in 3ß-HSD expression was amplified and reversed in the HGrC1 cells when FSH receptor expression was up-regulated by activin-A and down-regulated with shRNA, respectively. LIMITATIONS AND REASONS FOR CAUTION: As only the effect of FSH was studied we cannot extrapolate our findings to the potential effects of HMG and recombinant LH. WIDER IMPLICATIONS OF THE FINDINGS: This data provides a molecular explanation for the largely unexplained phenomenon of P rise during the follicular phase of gonadotropin stimulated IVF cycles. Our findings may progress the research to uncover potential mechanisms for preventing premature P rise that appears to be associated with inferior outcomes in women undergoing IVF. STUDY FUNDING/COMPETING INTEREST(S): Funded by the School of Medicine and the Graduate School of Health Sciences of Koc University. All authors declare no conflict of interest. TRIAL REGISTRATION NUMBER: None.


Assuntos
Hormônio Foliculoestimulante/uso terapêutico , Células da Granulosa/efeitos dos fármacos , Luteinização/efeitos dos fármacos , Progesterona/biossíntese , 3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/metabolismo , Linhagem Celular , Feminino , Humanos , Indução da Ovulação/métodos , Pregnenolona/metabolismo , Progesterona/sangue , RNA Mensageiro/metabolismo , Regulação para Cima/efeitos dos fármacos
10.
J Reprod Dev ; 63(1): 75-85, 2017 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-27840375

RESUMO

Hypoxia has been suggested to enhance progesterone (P4) synthesis in luteinizing granulosa cells (GCs), but the mechanism is unclear. The present study was designed to test the hypothesis that the hypoxia-induced increase in P4 synthesis during luteinization in bovine GCs is mediated by hypoxia-inducible factor 1 (HIF-1). GCs obtained from small antral follicles were cultured with 2 µg/ml insulin in combination with 10 µM forskolin for 24 h as a model of luteinizing GCs. To examine the influence of HIF-1 on P4 synthesis, we determined the effect of changes in protein expression of the α-subunit of HIF-1 (HIF1A) on P4 production and on the expression levels of StAR, P450scc, and 3ß-HSD. CoCl2 (100 µM), a hypoxia-mimicking chemical, increased HIF-1α protein expression in luteinizing GCs. After the upregulation of HIF-1α, we observed an increase in P4 production and in the gene and protein expression levels of StAR in CoCl2-treated luteinizing GCs. In contrast, CoCl2 did not affect the expression of either P450scc or 3ß-HSD. Echinomycin, a small-molecule inhibitor of HIF-1's DNA-binding activity, attenuated the effects of CoCl2 and of low oxygen tension (10% O2) on P4 production and StAR expression in luteinizing GCs. Overall, these findings suggest that HIF-1 is one of the factors that upregulate P4 in GCs during luteinization.


Assuntos
Células da Granulosa/citologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Luteinização/efeitos dos fármacos , Progesterona/biossíntese , Animais , Bovinos , Sobrevivência Celular , Colforsina/metabolismo , DNA/química , DNA Complementar/metabolismo , Equinomicina/química , Feminino , Células da Granulosa/metabolismo , Hipóxia , Hormônio Luteinizante/metabolismo , Folículo Ovariano/metabolismo , Ovário/metabolismo , Oxigênio/metabolismo , RNA Mensageiro/metabolismo , Ativação Transcricional , Regulação para Cima
11.
J Pineal Res ; 61(3): 279-90, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27177022

RESUMO

The functions of melatonin in preovulatory fluid remain elusive. In the current study, we observed that the extremely high level of expression of MT1 in mice granulosa cells was rapidly induced by hCG (equivalent LH) within 2 hours and this was referred as MT1 surge. In cumulus cells, serotonin N-acetyltransferase (SNAT) was also upregulated by hCG and led to elevated melatonin levels in ovarian follicle fluid. Melatonin application before MT1 surge significantly promoted embryo implantation, and this was probably attributed to a rise in progesterone levels in the serum. The mechanistic studies indicated that melatonin/MT1 (MLT/MT1) signaling remarkably improved the expression of corpus luteum marker genes, that is, Akr1c18 and Cyp11a1. High-throughput sequencing results suggested that extracellular matrix (ECM) receptor interaction, focal adhesion, and activation of PI3K/Akt pathway which are involved in granulosa cell luteinization might mediate the actions of MLT/MT1 signal. In addition, this effect on luteinization was compared in different species. It was verified that high melatonin levels exist in serum at estrum of cows and help to improve the first estrus fecundation rate. These results suggested that both melatonin and MT1 are involved in the downstream reaction of hCG (LH) and they play important roles in luteinization. These findings provide the novel information on the physiology of melatonin in animal reproduction.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/metabolismo , Luteinização/efeitos dos fármacos , Hormônio Luteinizante/metabolismo , Melatonina/farmacologia , Receptor MT1 de Melatonina/biossíntese , Transdução de Sinais/efeitos dos fármacos , Animais , Feminino , Masculino , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
12.
Biol Reprod ; 94(5): 109, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26985003

RESUMO

Adenoviral vectors (vectors) expressing short-hairpin RNAs complementary to macaque nuclear progesterone (P) receptor PGR mRNA (shPGR) or a nontargeting scrambled control (shScram) were used to determine the role PGR plays in ovulation/luteinization in rhesus monkeys. Nonluteinized granulosa cells collected from monkeys (n = 4) undergoing controlled ovarian stimulation protocols were exposed to either shPGR, shScram, or no virus for 24 h; human chorionic gonadotropin (hCG) was then added to half of the wells to induce luteinization (luteinized granulosa cells [LGCs]; n = 4-6 wells/treatment/monkey). Cells/media were collected 48, 72, and 120 h postvector for evaluation of PGR mRNA and P levels. Addition of hCG increased (P < 0.05) PGR mRNA and medium P levels in controls. However, a time-dependent decline (P < 0.05) in PGR mRNA and P occurred in shPGR vector groups. Injection of shPGR, but not shScram, vector into the preovulatory follicle 20 h before hCG administration during controlled ovulation protocols prevented follicle rupture in five of six monkeys as determined by laparoscopic evaluation, with a trapped oocyte confirmed in three of four follicles of excised ovaries. Injection of shPGR also prevented the rise in serum P levels following the hCG bolus compared to shScram (P < 0.05). Nuclear PGR immunostaining was undetectable in granulosa cells from shPGR-injected follicles, compared to intense staining in shScram controls. Thus, the nuclear PGR appears to mediate P action in the dominant follicle promoting ovulation in primates. In vitro and in vivo effects of PGR knockdown in LGCs also support the hypothesis that P enhances its own synthesis in the primate corpus luteum by promoting luteinization.


Assuntos
Células da Granulosa/metabolismo , Ovulação/genética , Progesterona/metabolismo , Receptores de Progesterona/genética , Animais , Células Cultivadas , Corpo Lúteo/efeitos dos fármacos , Corpo Lúteo/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Células Lúteas/efeitos dos fármacos , Células Lúteas/metabolismo , Luteinização/efeitos dos fármacos , Luteinização/genética , Macaca mulatta , Progesterona/farmacologia , Transfecção
13.
Biol Reprod ; 93(6): 133, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26510866

RESUMO

The surge of luteinizing hormone triggers the genomic reprogramming, cell differentiation, and tissue remodeling of the ovulated follicle, leading to the formation of the corpus luteum. During this process, called luteinization, follicular granulosa cells begin expressing a new set of genes that allow the resulting luteal cells to survive in a vastly different hormonal environment and to produce the extremely high amounts of progesterone (P4) needed to sustain pregnancy. To better understand the molecular mechanisms involved in the regulation of luteal P4 production in vivo, the transcription factors GATA4 and GATA6 were knocked down in the corpus luteum by crossing mice carrying Gata4 and Gata6 floxed genes with mice carrying Cre recombinase fused to the progesterone receptor. This receptor is expressed exclusively in granulosa cells after the luteinizing hormone surge, leading to recombination of floxed genes during follicle luteinization. The findings demonstrated that GATA4 and GATA6 are essential for female fertility, whereas targeting either factor alone causes subfertility. When compared to control mice, serum P4 levels and luteal expression of key steroidogenic genes were significantly lower in conditional knockdown mice. The results also showed that GATA4 and GATA6 are required for the expression of the receptors for prolactin and luteinizing hormone, the main luteotropic hormones in mice. The findings demonstrate that GATA4 and GATA6 are crucial regulators of luteal steroidogenesis and are required for the normal response of luteal cells to luteotropins.


Assuntos
Corpo Lúteo/metabolismo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA6/genética , Infertilidade Feminina/genética , Luteinização/genética , Progesterona/biossíntese , Animais , Gonadotropina Coriônica/farmacologia , Corpo Lúteo/efeitos dos fármacos , Feminino , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA6/metabolismo , Técnicas de Silenciamento de Genes , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Infertilidade Feminina/metabolismo , Luteinização/efeitos dos fármacos , Luteinização/metabolismo , Camundongos , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
14.
PLoS One ; 9(10): e109133, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25299186

RESUMO

BACKGROUND: Recent studies have indicated the use of gonadotropin-releasing hormone antagonists (GnRH-ant) as an adjuvant treatment to prevent premature luteinization (PL) and improve the clinical outcomes in patients undergoing controlled ovarian stimulation (COS) with intrauterine insemination (IUI). However, the results of these studies are conflicting. METHODS: We conducted a systematic review and meta-analysis of randomized trials aiming to compare the clinical efficacy of GnRH-ant in COS/IUI cycles. Twelve studies were identified that met inclusion criteria and comprised 2,577 cycles assigned to COS/IUI combined GnRH-ant or COS/IUI alone. RESULTS: Meta-analysis results suggested that GnRH-ant can significantly increase the clinical pregnancy rate (CPR) (OR = 1.42; 95% CI, 1.13-1.78) and decrease the PL rate (OR = 0.22, 95% CI, 0.16-0.30) in COS/IUI cycles. Subgroup analysis results suggested statistically significant improvement in the CPR in non-PCOS patients (OR = 1.54; 95% CI, 1.03-2.31) but not in the PCOS population (OR = 1.65; 95% CI, 0.93-2.94) and multiple mature follicle cycles (OR = 1.87; 95% CI, 0.27-12.66). There were no difference in the miscarriage and multiple pregnancy rates between the groups. CONCLUSION: This meta-analysis suggested that GnRH-ant can reduce the incidence of PL and increase the CPR when used in COS/IUI cycles, and it was especially useful for non-PCOS patients. However, evidence to support its use in PCOS patients is still insufficient.


Assuntos
Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Antagonistas de Hormônios/uso terapêutico , Infertilidade/tratamento farmacológico , Adolescente , Adulto , Feminino , Fertilização in vitro/métodos , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Humanos , Infertilidade/metabolismo , Inseminação Artificial/métodos , Luteinização/efeitos dos fármacos , Masculino , Indução da Ovulação/métodos , Gravidez , Taxa de Gravidez , Adulto Jovem
15.
Biol Reprod ; 91(5): 124, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25253736

RESUMO

Premature luteinization is a possible cause of infertility in women. It is currently unknown whether environmental chemicals can induce changes associated with premature luteinization. Using rat granulosa cells (GC) in vitro, we demonstrated that exposure to atrazine (ATR), a widely used herbicide, causes GC phenotype that resembles that of human premature luteinization. At the end of the 48-h stimulation with FSH, ATR-exposed GC showed (1) higher levels of progesterone, (2) overexpression of luteal markers (Star and Cyp11a1), and (3) an increase in progesterone:estradiol ratio above 1. Mechanistic experiments were conducted to understand the signaling events engaged by ATR that lead to this phenotype. Western blot analysis revealed prolonged phosphorylation of protein kinase B (AKT) and cAMP response element-binding protein (CREB) in ATR- and FSH-exposed GC. An increased level of ERK1/2-dependent transcriptional factor CCATT/enhancer-binding protein beta (CEBPB) was observed after 4 h of ATR exposure. Inhibitors of PI3K (wortmannin) and MEK (U0126) prevented ATR-induced rise in progesterone level and expression of luteal markers in FSH-stimulated GC. Atrazine intensified AKT and CEBPB signaling and caused Star overexpression in forskolin-stimulated GC but not in epidermal growth factor (EGF)-stimulated GC. In the presence of rolipram, a specific inhibitor of phosphodiesterase 4 (PDE4), ATR was not able to further elevate AKT phosphorylation, CEBPB protein level, and Star mRNA in FSH-stimulated GC, suggesting that ATR inhibits PDE4. Overall, this study showed that ATR acts as a FSH sensitizer leading to enhanced cAMP, AKT, and CEBPB signaling and progesterone biosynthesis, which promotes premature luteinization phenotype in GC.


Assuntos
Atrazina/farmacologia , Hormônio Foliculoestimulante/farmacologia , Células da Granulosa/efeitos dos fármacos , Herbicidas/farmacologia , Luteinização/efeitos dos fármacos , Progesterona/metabolismo , Animais , Células Cultivadas , Feminino , Células da Granulosa/metabolismo , Luteinização/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
16.
J Reprod Dev ; 60(3): 194-201, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24583842

RESUMO

To determine whether hypoxia has an effect on luteinization, we examined the influence of hypoxia on a model of bovine luteinizing and non-luteinizing granulosa cell culture. The granulosa cells were obtained from small antral follicles (≤ 6 mm in diameter). To induce luteinization, the cells were treated for 24 h with insulin (2 µg/ml), forskolin (10 µM) or insulin in combination with forskolin at 20% O2. After 24 h, progesterone (P4) production was higher in the treated cells, which we defined as luteinizing granulosa cells, than in non-treated cells, which we defined as non-luteinizing granulosa cells. P4 production by non-luteinizing granulosa cells was not affected by hypoxia (24 h at 10% and 5% O2), while P4 production by granulosa cells treated with insulin in combination with forskolin was significantly increased under hypoxia (24 h at 10% and 5% O2). Because hypoxia affected P4 production by the luteinizing granulosa cells but not by the non-luteinizing granulosa cells, hypoxia seems to promote P4 production during, rather than before, luteinization. In the cells treated with insulin in combination with forskolin, mRNA and protein expression of steroidogenic acute regulatory protein (StAR) and protein expression of 3ß-hydroxysteroid dehydrogenase (3ß-HSD) increased under 10% O2, while mRNA and protein expressions of key protein and enzymes in P4 biosynthesis did not increase under 5% O2. The overall results suggest that hypoxia plays a role in progressing and completing the luteinization by enhancing P4 production through StAR as well as 3ß-HSD expressions in the early time of establishing the corpus luteum.


Assuntos
Células da Granulosa/metabolismo , Hipóxia/metabolismo , Luteinização/metabolismo , Progesterona/biossíntese , 3-Hidroxiesteroide Desidrogenases/genética , 3-Hidroxiesteroide Desidrogenases/metabolismo , Animais , Bovinos , Células Cultivadas , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Luteinização/efeitos dos fármacos , Oxigênio/farmacologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo
17.
J Steroid Biochem Mol Biol ; 143: 90-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24607812

RESUMO

Cultured ovarian granulosa cells (GCs) are essential models to study molecular mechanisms of gene regulation during folliculogenesis. CCAAT enhancer binding proteins ß (CEBPß) has been identified in the ovary and is critical for follicular growth, ovulation and luteinization in mice. In the present study, hormonal treatment indicated that luteinizing hormone (LH) and exogenous human chorionic gonadotropins (hCG) significantly increased the expression of CEBPß in porcine GCs. By RNAi-Ready pSIREN-RetroQ-ZsGreen Vector mediated recombinant pshRNA vectors, CEBPß gene was successfully knocked down in porcine GCs, confirmed by mRNA and protein level analyzed by real time PCR and western blot, respectively. We further found that knockdown of CEBPß significantly increased the expression of p-ERK1/2. Furthermore, CEBPß knockdown arrested the GCs at S phase of cell cycle, but had no effects on cell apoptosis. More importantly, it markedly down regulated the concentration of estradiol (E2) and progesterone (P4) in the culture medium. To uncover the regulatory mechanism of CEBPß knockdown on cell cycle and steroids synthesis, we found that the mRNA expression of bcl-2 (anti-apoptosis), StAR and Runx2 (steroid hormone synthesis) was up-regulated, while genes related to apoptosis (Caspase-3 and p53), hormonal synthesis (CYP11A1) and cell cycle (cyclinA1, cyclinB1, cyclinD1) were down-regulated, suggesting that knockdown of CEBPß may inhibit apoptosis, regulate cell cycle and hormone secretions at the transcriptional level in porcine GCs. Furthermore, knockdown of CEBPß significantly increased the expression of PTGS2 and decreased the expression of IGFBP4, Has2 and PTGFR which are important for folliculogenesis in porcine GCs. In conclusion, this study reveals that CEBPß is a key regulator of porcine GCs through modulation of cell cycle, apoptosis, steroid synthesis, and other regulators of folliculogenesis.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/antagonistas & inibidores , Pontos de Checagem do Ciclo Celular/fisiologia , Estradiol/metabolismo , Células da Granulosa/metabolismo , Progesterona/metabolismo , RNA Interferente Pequeno/genética , Fase S/fisiologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Gonadotropina Coriônica/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Regulação da Expressão Gênica , Células da Granulosa/citologia , Células da Granulosa/efeitos dos fármacos , Luteinização/efeitos dos fármacos , Hormônio Luteinizante/farmacologia , Ovulação/efeitos dos fármacos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Substâncias para o Controle da Reprodução/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fase S/efeitos dos fármacos , Suínos
18.
Gen Comp Endocrinol ; 200: 10-7, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24556528

RESUMO

The ovarian promoter of the primate and rodent genes encoding cytochrome P450 aromatase (CYP19A1) are robustly responsive to forskolin in luteinized cell models, whereas the ruminant ovarian promoter is minimally active. We explored this discrepancy by investigating the activity of the bovine ovarian promoter in two bovine granulosa cell models, luteinizing and non-luteinizing cells in vitro. In non-luteinizing cells, both FSH and IGF1 increased abundance of transcripts derived from the ovarian promoter. Comparison of the activity of promoters of several species in response to transcription factors (forskolin, NR5A2, FOXL2) in luteinizing cells demonstrated that a rat ovarian promoter-luciferase reporter was regulated mainly by forskolin (18-fold increase over basal expression) and addition of NR5A2 or FOXL2 had no further effect. Activity of a human promoter was significantly increased by NR5A2 plus forskolin (153-fold) compared with forskolin alone (71-fold over basal); addition of FOXL2 did not significantly increase promoter activity. Forskolin alone provoked minor activation of caprine and bovine promoter reporters (3-fold over basal), and addition of NR5A2 increased activity (7- to 11-fold). When forskolin, NR5A2 and FOXL2 treatments were combined, the activity of the caprine and bovine promoters increased to 20- and 34-fold, respectively. These data suggest that a major reason why CYP19A1 is not expressed in luteinized cells (and the corpus luteum) of ruminants may be the stimulatory effect of FOXL2, which does not appear to be the case in the human and rat.


Assuntos
Aromatase/genética , Células da Granulosa/enzimologia , Modelos Biológicos , Regiões Promotoras Genéticas , Animais , Aromatase/metabolismo , Bovinos , Colforsina/farmacologia , Corpo Lúteo/metabolismo , Estradiol/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Hormônio Foliculoestimulante/farmacologia , Fatores de Transcrição Forkhead/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Cabras , Células da Granulosa/efeitos dos fármacos , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Luteinização/efeitos dos fármacos , Luteinização/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores do FSH/metabolismo , Especificidade da Espécie , Transfecção
19.
Reprod Fertil Dev ; 26(3): 367-74, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23458081

RESUMO

This study examined the effect of a single administration of human chorionic gonadotrophin (hCG) on Day 1 to 4 after oestrus on corpus luteum (CL) development and circulating progesterone (P4). Oestrus-synchronized heifers (n=43) were administered a single intramuscular injection of saline on Day 1 (control) or 3000IU hCG on Day 1, 2, 3 or 4 after oestrus. Administration of hCG on Day 1 had no effect on CL area, on Day 2 increased CL area from Day 6 to 12 (P<0.05), on Day 3 increased CL area from Day 9 to 11, while on Day 4 increased CL size on Days 9 and 10 (P<0.05). Administration of hCG on Day 4 induced the formation of an accessory CL in 89% of heifers, resulting in a significant increase in total luteal tissue area on the ovaries compared with all other groups. Consistent with the effects on the CL, hCG on Day 1 did not affect P4 concentrations, on Day 2 significantly increased P4 compared with the control from Day 6 to 11 (P<0.05), on Day 3 resulted in a non-significant increase in P4 while hCG on Day 4 increased P4 from Day 8 to 13 compared with the control (P<0.05). In conclusion, administration of hCG as early as Day 2 after oestrus results in increased P4 in circulation from Day 6, which should have beneficial downstream effects in terms of uterine receptivity and conceptus elongation.


Assuntos
Gonadotropina Coriônica/farmacologia , Corpo Lúteo/crescimento & desenvolvimento , Luteinização/fisiologia , Progesterona/sangue , Animais , Cruzamento/métodos , Bovinos , Corpo Lúteo/diagnóstico por imagem , Corpo Lúteo/efeitos dos fármacos , Sincronização do Estro , Feminino , Humanos , Luteinização/efeitos dos fármacos , Modelos Estatísticos , Ultrassonografia
20.
Reprod Sci ; 21(4): 444-55, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24077439

RESUMO

Excess of prenatal testosterone (T) induces reproductive defects including follicular persistence. Comparative studies with T and dihydrotestosterone (DHT) have suggested that follicular persistence is programmed via estrogenic actions of T. This study addresses the androgenic and estrogenic contributions in programming follicular persistence. Because humans are exposed to estrogenic environmental steroids from various sources throughout their life span and postnatal insults may also induce organizational and/or activational changes, we tested whether continuous postnatal exposure to estradiol (E) will amplify effects of prenatal steroids on ovarian function. Pregnant sheep were treated with T, DHT, E, or ED (E and DHT) from days 30 to 90 of gestation. Postnatally, a subset of the vehicle (C), T, and DHT females received an E implant. Transrectal ultrasonography was performed in the first breeding season during a synchronized cycle to monitor ovarian follicular dynamics. As expected, number of ≥8 mm follicles was higher in the T versus C group. Postnatal E reduced the number of 4 to 8 mm follicles in the DHT group. Percentage of females bearing luteinized follicles and the number of luteinized follicles differed among prenatal groups. Postnatal E increased the incidence of subluteal cycles in the prenatal T-treated females. Findings from this study confirm previous findings of divergences in programming effects of prenatal androgens and estrogens. They also indicate that some aspects of follicular dynamics are subject to postnatal modulation as well as support the existence of an extended organizational period or the need for a second insult to uncover the previously programmed event.


Assuntos
Di-Hidrotestosterona/toxicidade , Estradiol/toxicidade , Folículo Ovariano/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Testosterona/toxicidade , Animais , Implantes de Medicamento , Estradiol/administração & dosagem , Ciclo Estral/sangue , Ciclo Estral/efeitos dos fármacos , Feminino , Idade Gestacional , Luteinização/efeitos dos fármacos , Folículo Ovariano/diagnóstico por imagem , Folículo Ovariano/metabolismo , Gravidez , Progesterona/sangue , Ovinos , Fatores de Tempo , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA