Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Reprod Fertil Dev ; 33(16): 810-816, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34758896

RESUMO

Atypical protein serine kinase RIOK3 is involved in cellular invasion and survival. The spatiotemporal expression pattern and regulatory mechanisms controlling expression of Riok3 were investigated in the rat ovary during the periovulatory period. Immature female rats (22-23 days old) were treated with pregnant mare's serum gonadotropin (PMSG) to stimulate follicular development, followed 48h later by injection with human chorionic gonadotrophin (hCG). Ovaries, granulosa cells, or theca-interstitial cells were collected at various times after hCG administration. Both real-time polymerase chain reaction (PCR) and in situ hybridisation analysis revealed that Riok3 was highly induced in both granulosa cells and theca-interstitial cells by hCG. Riok3 expression was induced in theca-interstitial cells at 4h after hCG. However, the expression of Riok3 mRNA was stimulated in granulosa cells at 8h. Both protein kinase C inhibitor (GF109203) and the protein kinase A inhibitor (H89) could block the stimulation of Riok3 mRNA by hCG. Furthermore, Riok3 induction is dependent on new protein synthesis. Inhibition of prostaglandin synthesis or progesterone action did not alter Riok3 mRNA expression, whereas inhibition of the epidermal growth factor (EGF) pathway downregulated Riok3 expression. In conclusion, our findings suggest that the induction of the RIOK3 may be important for ovulation and luteinisation.


Assuntos
Luteinização/metabolismo , Ovário/metabolismo , Ovulação/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Feminino , Gonadotropinas Equinas/farmacologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Luteinização/efeitos dos fármacos , Luteinização/genética , Ovário/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Ovulação/genética , Proteínas Serina-Treonina Quinases/genética , Ratos , Transdução de Sinais/efeitos dos fármacos
2.
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
3.
Endocrinology ; 162(9)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34171084

RESUMO

The ovulatory luteinizing hormone (LH) surge induces rapid changes of gene expression and cellular functions in granulosa cells (GCs) undergoing luteinization. However, it remains unclear how the changes in genome-wide gene expression are regulated. H3K4me3 histone modifications are involved in the rapid alteration of gene expression. In this study, we investigated genome-wide changes of transcriptome and H3K4me3 status in mouse GCs undergoing luteinization. GCs were obtained from mice treated with equine chorionic gonadotropin (hCG) before, 4 hours, and 12 hours after human chorionic gonadotropin injection. RNA-sequencing identified a number of upregulated and downregulated genes, which could be classified into 8 patterns according to the time-course changes of gene expression. Many genes were transiently upregulated or downregulated at 4 hours after hCG stimulation. Gene Ontology terms associated with these genes included steroidogenesis, ovulation, cumulus-oocyte complex (COC) expansion, angiogenesis, immune system, reactive oxygen species (ROS) metabolism, inflammatory response, metabolism, and autophagy. The cellular functions of DNA repair and cell growth were newly identified as being activated during ovulation. Chromatin immunoprecipitation-sequencing revealed a genome-wide and rapid change in H3K4me3 during ovulation. Integration of transcriptome and H3K4me3 data identified many H3K4me3-associated genes that are involved in steroidogenesis, ovulation, COC expansion, angiogenesis, inflammatory response, immune system, ROS metabolism, lipid and glucose metabolism, autophagy, and regulation of cell size. The present results suggest that genome-wide changes in H3K4me3 after the LH surge are associated with rapid changes in gene expression in GCs, which enables GCs to acquire a lot of cellular functions within a short time that are required for ovulation and luteinization.


Assuntos
Células da Granulosa/metabolismo , Histonas/metabolismo , Ovulação/fisiologia , Transcriptoma , Animais , Gonadotropina Coriônica/farmacologia , Feminino , Perfilação da Expressão Gênica , Células da Granulosa/efeitos dos fármacos , Código das Histonas/efeitos dos fármacos , Código das Histonas/genética , Luteinização/efeitos dos fármacos , Luteinização/genética , Luteinização/metabolismo , Hormônio Luteinizante/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ovulação/genética , Ovulação/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Integração de Sistemas , Transcriptoma/efeitos dos fármacos
4.
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
5.
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
6.
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
7.
Mol Cell Endocrinol ; 479: 87-92, 2019 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-30253183

RESUMO

Vitamin D3 (1,25-dihydroxyvitamin D3, VD3) in vitro attenuates the effect of the pro-inflammatory advanced glycation end products (AGEs) on steroidogenesis in human granulosa cells (GCs) by downregulating the receptor for AGEs (RAGE). It has been shown that VD3 alone downregulates anti-Mullerian hormone (AMH) type 2 receptor (AMHR-2) gene expression and suppresses AMH-induced SMAD 1/5/8 phosphorylation in granulosa cells. However, the effect of AGEs, in the absence or presence of VD3, on AMH action in GCs has not been studied. Using human GCs, this study showed that human glycated albumin (HGA), an in vitro representative for AGEs, upregulated AMHR-2 mRNA but did not alter AMH mRNA expression levels. VD3 inhibited the HGA-induced increase in AMHR-2 mRNA expression levels. In KGN granulosa cell line, recombinant AMH induced SMAD 1/5/8 phosphorylation. HGA augmented the recombinant AMH-induced SMAD 1/5/8 phosphorylation while the addition of VD3 to HGA attenuated the recombinant AMH-induced SMAD 1/5/8 phosphorylation. Thus, AGEs could potentially affect folliculogenesis as reflected by changes in AMH signaling. These findings have significant implications for women with polycystic ovary syndrome who have significantly elevated serum and ovarian AGEs.


Assuntos
Hormônio Antimülleriano/metabolismo , Produtos Finais de Glicação Avançada/toxicidade , Transdução de Sinais , Vitamina D/farmacologia , Adulto , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/metabolismo , Humanos , Luteinização/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Recombinantes/farmacologia , Albumina Sérica/toxicidade , Proteínas Smad/metabolismo , Albumina Sérica Glicada
8.
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
9.
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
10.
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
11.
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
12.
Anim Sci J ; 88(11): 1686-1691, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28594136

RESUMO

Bone morphogenetic protein-4 (BMP-4) inhibits luteinization of granulosa cells during in vitro growth (IVG) culture of bovine oocytes; however, oocytes derived from a 12 day IVG were less competent for development than in vivo-grown oocytes. We herein investigated whether an extended IVG culture with BMP-4 improves oocyte growth and development to blastocysts after in vitro fertilization. Oocyte-granulosa cell complexes (OGCs) were cultured for 14 or 16 days with BMP-4 (10 ng/mL), while a 12 day culture with BMP-4 served as the in vitro control. OGC viability was maintained for the 16 day culture with BMP-4 (83.2%), but was significantly lower without BMP-4 (58.9%) than the control (83.0%). Prolong-cultured oocytes at 16 days had statistically greater diameter (114.6 µm) than the control (111.7 µm). IVG oocytes with BMP-4 for the 16 day culture had a similar nuclear maturation rate to the control (approximately 67%); however, blastocyst rates in BMP-4 treated oocytes of 14 (1.8%) and 16 day (0%) IVG were statistically lower than that of 12 day IVG (9.0%). In conclusion, BMP-4 maintained OGC viability and promoted oocyte growth in a prolonged culture, but impaired the developmental competence of oocytes. Prolonged culture may not be an appropriate strategy for enhancing the developmental competence of IVG oocytes.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Oócitos/crescimento & desenvolvimento , Animais , Blastocisto , Bovinos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Fertilização in vitro , Células da Granulosa/fisiologia , Luteinização/efeitos dos fármacos , Fatores de Tempo
13.
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
14.
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
15.
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
16.
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
17.
Equine Vet J ; 48(2): 211-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25438830

RESUMO

REASONS FOR PERFORMING STUDY: Haemorrhagic anovulatory follicle is the most common pathological anovulatory condition in the mare, but its cause remains unknown. An experimental model to induce luteinised unruptured follicles (LUF) with flunixin-meglumine (FM) has been developed. Luteinised unruptured follicles share similar morphological and hormonal characteristics with haemorrhagic anovulatory follicles. OBJECTIVES: To test the effect of intrafollicular administration of prostaglandins PGE2 and PGF2α during the periovulatory period on ovulation and pregnancy in FM-treated mares. STUDY DESIGN: In vivo experiment in a crossover design. METHODS: Five mares were followed during 2 oestrous cycles each. All mares were given FM at 1.7 mg/kg bwt i.v. every 12 h from Hour 0 (Hour 0 = human chorionic gonadotrophin treatment) to Hour 36. In treatment cycles (n = 5), at Hour 32 the preovulatory follicle was punctured and 0.5 ml of a solution containing 500 µg of PGE2 and 125 µg of PGF2α was deposited within the follicle. In control cycles, water for injection was administered into the follicle at the same time. In 3 control and 3 treatment cycles, mares were also inseminated at Hour 24. Diagnosis of ovulation/LUF formation and pregnancy was performed by ultrasound examination between Hours 36 and 72 and 14 days after ovulation/LUF formation, respectively. RESULTS: During the treatment cycles, all mares ovulated normally (100% ovulation rate) 36-48 h after human chorionic gonadotrophin, while in 4 of 5 control cycles the mares developed an LUF (80%, P<0.05). All 3 inseminated mares became pregnant in the treatment cycles, but not in the control cycles. CONCLUSIONS: Intrafollicular treatment with PGE2 and PGF2α overcame the anovulatory effect of FM. This sheds new insights into the knowledge on the possible therapeutic options for ovulatory failure in the mare.


Assuntos
Clonixina/análogos & derivados , Dinoprosta/farmacologia , Dinoprostona/farmacologia , Luteinização/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Ovulação/efeitos dos fármacos , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacologia , Clonixina/farmacologia , Dinoprostona/administração & dosagem , Feminino , Cavalos , Gravidez
18.
J Zhejiang Univ Sci B ; 16(12): 980-90, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26642181

RESUMO

The effects of equine chorionic gonadotropin (eCG) on follicular development and ovulation in cyclic guinea pigs were investigated by histological and immunohistochemical analyses. Three groups of guinea pigs (n=12) were administrated subcutaneously with saline, 20 or 50 IU of eCG, respectively, on cyclic Day 12 (Day 1=vaginal openings). Ovaries were collected at 4 and 8 d after administration (6 animals per group each time). The eCG administration induced significant and distinct morphological changes in the ovaries, as it promoted the luteinization of granulosa cells, but not follicular development. In addition, proliferating cell nuclear antigen (PCNA) and steroidogenic acute regulatory protein (StAR) were immunolocalized specifically in luteinized follicles. Our experiments together indicate that eCG administration can induce follicular luteinization but not superovulation in guinea pigs. The eCG in cyclic guinea pigs functions similar to that of luteinizing hormone (LH), but not follicle-stimulating hormone (FSH).


Assuntos
Gonadotropinas Equinas/administração & dosagem , Luteinização/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Atresia Folicular/efeitos dos fármacos , Cobaias , Imuno-Histoquímica , Luteinização/fisiologia , Oócitos/citologia , Oócitos/efeitos dos fármacos , Folículo Ovariano/citologia , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/fisiologia , Fosfoproteínas/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Superovulação/efeitos dos fármacos
19.
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
20.
Reprod Domest Anim ; 50(5): 771-5, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26280558

RESUMO

The presence of anovulatory haemorrhagic follicles during the oestrous cycle of mares causes financial impacts, slowing conception and increasing the number of services per pregnancy. Non-steroidal anti-inflammatory drugs (NSAIDs) such as meloxicam and phenylbutazone are used in the treatment of several disorders in mares, and these drugs can impair the formation of prostaglandins (PGs) and consequently interfere with reproductive activity. This study aimed to evaluate the effects of treatment with NSAIDs on the development of pre-ovulatory follicles in mares. In total, 11 mares were studied over three consecutive oestrous cycles, and gynaecological and ultrasound examinations were performed every 12 h. When 32-mm-diameter follicles were detected, 1 mg of deslorelin was administered to induce ovulation. The first cycle was used as a control, and the mares received only a dose of deslorelin. In the subsequent cycles, in addition to receiving the same dose of deslorelin, each mare was treated with NSAIDs. In the second cycle, 4.4 mg/kg of phenylbutazone was administered, and in the third cycle, 0.6 mg/kg of meloxicam was administered once a day until ovulation or the beginning of follicular haemorrhage. All of the mares ovulated between 36 and 48 h after the induction in the control cycle. In the meloxicam cycle, 10 mares (92%) did not ovulate, while in the phenylbutazone cycle, nine mares (83%) did not ovulate. In both treatments, intrafollicular hyperechoic spots indicative of haemorrhagic follicles were observed on ultrasound. Thus, our results suggested that treatment with meloxicam and phenylbutazone at therapeutic doses induced intrafollicular haemorrhage and luteinization of anovulatory follicles.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Cavalos/fisiologia , Ovulação/efeitos dos fármacos , Fenilbutazona/administração & dosagem , Tiazinas/administração & dosagem , Tiazóis/administração & dosagem , Animais , Ciclo Estral , Feminino , Fase Folicular , Luteinização/efeitos dos fármacos , Masculino , Meloxicam , Folículo Ovariano/diagnóstico por imagem , Indução da Ovulação/veterinária , Gravidez , Pamoato de Triptorrelina/administração & dosagem , Pamoato de Triptorrelina/análogos & derivados , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA