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

RESUMEN

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.


Asunto(s)
Luteinización/metabolismo , Ovario/metabolismo , Ovulación/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Femenino , Gonadotropinas Equinas/farmacología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Luteinización/efectos de los fármacos , Luteinización/genética , Ovario/efectos de los fármacos , Ovulación/efectos de los fármacos , Ovulación/genética , Proteínas Serina-Treonina Quinasas/genética , Ratas , Transducción de Señal/efectos de los fármacos
2.
Reprod Fertil Dev ; 31(4): 698-704, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30414623

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Proteína HMGA1a/metabolismo , Luteinización/metabolismo , Ovario/metabolismo , Ovulación/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/metabolismo , Femenino , Regulación de la Expresión Génica , Gonadotropinas Equinas/farmacología , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Proteína HMGA1a/genética , Luteinización/efectos de los fármacos , Luteinización/genética , Ovulación/efectos de los fármacos , Ovulación/genética , Ratas , Ratas Sprague-Dawley , Células Tecales/efectos de los fármacos , Células Tecales/metabolismo
3.
J Reprod Dev ; 64(2): 129-134, 2018 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-29249775

RESUMEN

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.


Asunto(s)
Gonadotropina Coriónica/farmacología , Fármacos para la Fertilidad Femenina/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/agonistas , Células de la Granulosa/efectos de los fármacos , Luteinización/efectos de los fármacos , Ovario/efectos de los fármacos , Animales , Anexina A5/agonistas , Anexina A5/genética , Anexina A5/metabolismo , Biomarcadores/sangre , Biomarcadores/metabolismo , Células Cultivadas , Femenino , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Gonadotropinas Equinas/farmacología , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Humanos , Inmunohistoquímica , Ovario/citología , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Progesterona/agonistas , Progesterona/antagonistas & inhibidores , Progesterona/biosíntesis , Progesterona/sangre , Ratas Wistar , Receptores de HFE/agonistas , Receptores de HFE/antagonistas & inhibidores , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HL/agonistas , Receptores de HL/genética , Receptores de HL/metabolismo
4.
Gynecol Endocrinol ; 34(3): 189-191, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28942696

RESUMEN

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.


Asunto(s)
Fertilización In Vitro/métodos , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Gonadotropinas/administración & dosificación , Antagonistas de Hormonas/administración & dosificación , Luteinización/efectos de los fármacos , Inducción de la Ovulación/métodos , Ovulación/efectos de los fármacos , Adulto , Endometrio/diagnóstico por imagen , Femenino , Humanos , Persona de Mediana Edad , Folículo Ovárico/diagnóstico por imagen , Ultrasonografía
5.
Hum Reprod ; 32(3): 643-652, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28158500

RESUMEN

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.


Asunto(s)
Hormona Folículo Estimulante/uso terapéutico , Células de la Granulosa/efectos de los fármacos , Luteinización/efectos de los fármacos , Progesterona/biosíntesis , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Línea Celular , Femenino , Humanos , Inducción de la Ovulación/métodos , Pregnenolona/metabolismo , Progesterona/sangre , ARN Mensajero/metabolismo , Regulación hacia Arriba/efectos de los fármacos
6.
J Reprod Dev ; 63(1): 75-85, 2017 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-27840375

RESUMEN

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.


Asunto(s)
Células de la Granulosa/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Luteinización/efectos de los fármacos , Progesterona/biosíntesis , Animales , Bovinos , Supervivencia Celular , Colforsina/metabolismo , ADN/química , ADN Complementario/metabolismo , Equinomicina/química , Femenino , Células de la Granulosa/metabolismo , Hipoxia , Hormona Luteinizante/metabolismo , Folículo Ovárico/metabolismo , Ovario/metabolismo , Oxígeno/metabolismo , ARN Mensajero/metabolismo , Activación Transcripcional , Regulación hacia Arriba
7.
Biol Reprod ; 94(5): 109, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26985003

RESUMEN

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.


Asunto(s)
Células de la Granulosa/metabolismo , Ovulación/genética , Progesterona/metabolismo , Receptores de Progesterona/genética , Animales , Células Cultivadas , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Células Lúteas/efectos de los fármacos , Células Lúteas/metabolismo , Luteinización/efectos de los fármacos , Luteinización/genética , Macaca mulatta , Progesterona/farmacología , Transfección
8.
J Pineal Res ; 61(3): 279-90, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27177022

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/metabolismo , Luteinización/efectos de los fármacos , Hormona Luteinizante/metabolismo , Melatonina/farmacología , Receptor de Melatonina MT1/biosíntesis , Transducción de Señal/efectos de los fármacos , Animales , Femenino , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
9.
Biol Reprod ; 93(6): 133, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26510866

RESUMEN

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.


Asunto(s)
Cuerpo Lúteo/metabolismo , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA6/genética , Infertilidad Femenina/genética , Luteinización/genética , Progesterona/biosíntesis , Animales , Gonadotropina Coriónica/farmacología , Cuerpo Lúteo/efectos de los fármacos , Femenino , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/metabolismo , Técnicas de Silenciamiento del Gen , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Infertilidad Femenina/metabolismo , Luteinización/efectos de los fármacos , Luteinización/metabolismo , Ratones , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
10.
J Reprod Dev ; 61(5): 439-48, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26155753

RESUMEN

The corpus luteum (CL) is a temporary endocrine gland producing a large amount of progesterone, which is essential for the establishment and maintenance of pregnancy. Galectin-1 is a ß-galactose-binding protein that can modify functions of membrane glycoproteins and is expressed in the CL of mice and women. However, the physiological role of galectin-1 in the CL is unclear. In the present study, we investigated the expression and localization of galectin-1 in the bovine CL and the effect of galectin-1 on cultured luteal steroidogenic cells (LSCs) with special reference to its binding to the glycans on vascular endothelial growth factor receptor-2 (VEGFR-2). Galectin-1 protein was highly expressed at the mid and late luteal stages in the membrane fraction of bovine CL tissue and was localized to the surface of LSCs in a carbohydrate-dependent manner. Galectin-1 increased the viability in cultured LSCs. However, the viability of LSCs was decreased by addition of ß-lactose, a competitive carbohydrate inhibitor of galectin-1 binding activity. VEGFR-2 protein, like galectin-1, is also highly expressed in the mid CL, and it was modified by multi-antennary glycans, which can be recognized by galectin-1. An overlay assay using biotinylated galectin-1 revealed that galectin-1 directly binds to asparagine-linked glycans (N-glycans) on VEGFR-2. Enhancement of LSC viability by galectin-1 was suppressed by a selective inhibitor of VEGFR-2. The overall findings suggest that galectin-1 plays a role as a survival factor in the bovine CL, possibly by binding to N-glycans on VEGFR-2.


Asunto(s)
Cuerpo Lúteo/metabolismo , Galectina 1/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Lúteas/metabolismo , Luteinización , Polisacáridos/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/agonistas , Animales , Animales Endogámicos , Unión Competitiva , Bovinos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cuerpo Lúteo/citología , Cuerpo Lúteo/efectos de los fármacos , Femenino , Galectina 1/antagonistas & inhibidores , Galectina 1/genética , Humanos , Lactosa/análogos & derivados , Lactosa/metabolismo , Células Lúteas/citología , Células Lúteas/efectos de los fármacos , Luteinización/efectos de los fármacos , Polisacáridos/química , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Propiedades de Superficie , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Reprod Domest Anim ; 50(5): 771-5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26280558

RESUMEN

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.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Caballos/fisiología , Ovulación/efectos de los fármacos , Fenilbutazona/administración & dosificación , Tiazinas/administración & dosificación , Tiazoles/administración & dosificación , Animales , Ciclo Estral , Femenino , Fase Folicular , Luteinización/efectos de los fármacos , Masculino , Meloxicam , Folículo Ovárico/diagnóstico por imagen , Inducción de la Ovulación/veterinaria , Embarazo , Pamoato de Triptorelina/administración & dosificación , Pamoato de Triptorelina/análogos & derivados , Ultrasonografía
12.
Biol Reprod ; 91(5): 124, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25253736

RESUMEN

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.


Asunto(s)
Atrazina/farmacología , Hormona Folículo Estimulante/farmacología , Células de la Granulosa/efectos de los fármacos , Herbicidas/farmacología , Luteinización/efectos de los fármacos , Progesterona/metabolismo , Animales , Células Cultivadas , Femenino , Células de la Granulosa/metabolismo , Luteinización/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
13.
Reprod Fertil Dev ; 26(3): 367-74, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23458081

RESUMEN

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.


Asunto(s)
Gonadotropina Coriónica/farmacología , Cuerpo Lúteo/crecimiento & desarrollo , Luteinización/fisiología , Progesterona/sangre , Animales , Cruzamiento/métodos , Bovinos , Cuerpo Lúteo/diagnóstico por imagen , Cuerpo Lúteo/efectos de los fármacos , Sincronización del Estro , Femenino , Humanos , Luteinización/efectos de los fármacos , Modelos Estadísticos , Ultrasonografía
14.
Gen Comp Endocrinol ; 200: 10-7, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24556528

RESUMEN

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.


Asunto(s)
Aromatasa/genética , Células de la Granulosa/enzimología , Modelos Biológicos , Regiones Promotoras Genéticas , Animales , Aromatasa/metabolismo , Bovinos , Colforsina/farmacología , Cuerpo Lúteo/metabolismo , Estradiol/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Hormona Folículo Estimulante/farmacología , Factores de Transcripción Forkhead/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Cabras , Células de la Granulosa/efectos de los fármacos , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Luteinización/efectos de los fármacos , Luteinización/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de HFE/metabolismo , Especificidad de la Especie , Transfección
15.
J Reprod Dev ; 60(3): 194-201, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24583842

RESUMEN

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.


Asunto(s)
Células de la Granulosa/metabolismo , Hipoxia/metabolismo , Luteinización/metabolismo , Progesterona/biosíntesis , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Bovinos , Células Cultivadas , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Luteinización/efectos de los fármacos , Oxígeno/farmacología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo
16.
Biol Reprod ; 89(4): 91, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23966322

RESUMEN

Normal pregnancy is supported by increased levels of progesterone (P4), which is secreted from ovarian luteal cells via enzymatic steps catalyzed by P450scc (CYP11A1) and HSD3B. The development and maintenance of corpora lutea during pregnancy, however, are less well understood. Here we used Cyp11a1 transgenic mice to delineate the steps of luteal cell differentiation during pregnancy. Cyp11a1 in a bacterial artificial chromosome was injected into mouse embryos to generate transgenic mice with transgene expression that recapitulated endogenous Cyp11a1 expression. Cyp11a1 transgenic females displayed reduced pregnancy rate, impaired implantation and placentation, and decreased litter size in utero, although they produced comparable numbers of blastocysts. The differentiation of transgenic luteal cells was delayed during early pregnancy as shown by the delayed activation of genes involved in steroidogenesis and cholesterol availability. Luteal cell mitochondria were elongated, and their numbers were reduced, with morphology and numbers similar to those observed in granulosa cells. Transgenic luteal cells accumulated lipid droplets and secreted less progesterone during early pregnancy. The progesterone level returned to normal on gestation day 9 but was not properly withdrawn at term, leading to delayed stillbirth. P4 supplementation rescued the implantation rates but not the ovarian defects. Thus, overexpression of Cyp11a1 disrupts normal development of the corpus luteum, leading to progesterone insufficiency during early pregnancy. Misregulation of the progesterone production in Cyp11a1 transgenic mice during pregnancy resulted in aberrant implantation, anomalous placentation, and delayed parturition.


Asunto(s)
Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/biosíntesis , Decidua/enzimología , Infertilidad Femenina/enzimología , Células Lúteas/metabolismo , Luteinización/metabolismo , Progesterona/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Mantenimiento del Cuerpo Lúteo/sangre , Mantenimiento del Cuerpo Lúteo/efectos de los fármacos , Mantenimiento del Cuerpo Lúteo/metabolismo , Cruzamientos Genéticos , Decidua/efectos de los fármacos , Decidua/metabolismo , Decidua/patología , Implantación del Embrión/efectos de los fármacos , Femenino , Terapia de Reemplazo de Hormonas , Infertilidad Femenina/tratamiento farmacológico , Infertilidad Femenina/metabolismo , Infertilidad Femenina/patología , Metabolismo de los Lípidos/efectos de los fármacos , Tamaño de la Camada/efectos de los fármacos , Células Lúteas/efectos de los fármacos , Células Lúteas/patología , Luteinización/sangre , Luteinización/efectos de los fármacos , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Placentación/efectos de los fármacos , Embarazo , Progesterona/deficiencia , Progesterona/uso terapéutico , Organismos Libres de Patógenos Específicos
17.
Trop Anim Health Prod ; 45(1): 293-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22802094

RESUMEN

The current study was aimed to establish the impact of progesterone supplementation (norgestomet progestagen) between days 4 to 10 post-ovulation on subsequent luteal profile and conception rate in buffaloes. The 28 Murrah buffaloes of second to fourth parity, having normal reproductive organs, were estrus synchronized by double PGF(2α) protocol at 11 days apart. The buffaloes were inseminated during mid- to late estrus and thereafter repeated at 24 h interval. The buffaloes were randomly assigned into two groups: (1) control (no treatment, n = 14) and (2) treatment group (CRESTAR ear implant, n = 14). The CRESTAR ear implant (3 mg, norgestomet progestagen) was inserted subcutaneous between days 4 to 10 post-ovulation. The ovaries were scanned at estrus and thereafter on days 4, 10, 16, 21, and 40 post-ovulation to examine the preovulatory follicle (POF) and corpus luteum (CL) diameter. Each ultasonography was followed by blood sample collection for analysis of plasma progesterone concentrations following ovulation. The conception rate was similar (p > 0.05) between treated and control buffaloes. The pregnant buffalo of the control group had larger (p < 0.05) POF diameter than nonpregnant counterparts. The CL diameter was similar (p > 0.05) in both treated and untreated control as well as in their pregnant and nonpregnant buffaloes of the respective groups. The plasma progesterone concentrations were higher (p < 0.05) in the treatment group on the day 10 post-ovulation as compared to the control buffaloes. It is concluded that norgestomet supplementation had no impact on conception rate and CL diameter but enhances the plasma progesterone concentrations following treatment in buffaloes.


Asunto(s)
Búfalos/fisiología , Fertilización/efectos de los fármacos , Luteinización/efectos de los fármacos , Pregnenodionas/farmacología , Animales , Suplementos Dietéticos , Dinoprost , Femenino , Folículo Ovárico/citología , Embarazo , Pregnenodionas/administración & dosificación , Progesterona/sangre , Ultrasonografía/veterinaria
18.
Biol Reprod ; 87(6): 137, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23100619

RESUMEN

As the only domesticated species known to exhibit both induced and spontaneous ovulation, the cat is a model for understanding the nuances of ovarian control. To explore ovarian sensitivity to exogenous gonadotropins and the influence of progestin priming, we conducted a study of queens that were down-regulated with oral progestin or allowed to cycle normally, followed by low or high doses of equine chorionic gonadotropin (eCG) and human chorionic gonadotropin (hCG). Our metrics included 1) fecal steroid metabolite profiles before and after ovulation induction, 2) laparoscopic examination of ovarian follicles and corpora lutea (CL) on Days 2 and 17 (Day 0 = hCG administration), and 3) ovariohysterectomy (Day 17) to assess CL progesterone concentrations, morphometrics, and histology. Reproductive tracts from time-matched, naturally mated queens (n = 6) served as controls. Every progestin-primed cat (n = 12) produced the desired response of morphologically similar, fresh CL (regardless of eCG/hCG dose) by Day 2, whereas 41.7% of unprimed counterparts (n = 12) failed to ovulate or had variable-aged CL suggestive of prior spontaneous ovulation (P < 0.05). The ovarian response to low, but not high, eCG/hCG was improved (P < 0.05) in primed compared to unprimed cats, indicating increased sensitivity to gonadotropin in the progestin-primed ovary. Progestin priming prevented hyperelevated fecal steroid metabolites and normalized CL progesterone capacity, but only when combined with low eCG/hCG. However, priming failed to prevent ancillary CL formation, smaller CL mass, or abnormal luteal cell density, which were common to all eCG/hCG-treated cats. Thus, the domestic cat exposed to eCG/hCG produces CL with structural and functional aberrations. These anomalies can be partially mitigated by progestin priming, possibly due to a protective effect of progestin associated with enhanced ovarian sensitivity to gonadotropins.


Asunto(s)
Gonadotropina Coriónica/farmacología , Cuerpo Lúteo/efectos de los fármacos , Resistencia a Medicamentos/efectos de los fármacos , Células Lúteas/efectos de los fármacos , Inducción de la Ovulación/veterinaria , Ovulación/efectos de los fármacos , Progestinas/farmacología , Administración Oral , Animales , Gatos , Gonadotropina Coriónica/administración & dosificación , Gonadotropina Coriónica/efectos adversos , Cuerpo Lúteo/citología , Cuerpo Lúteo/metabolismo , Relación Dosis-Respuesta a Droga , Heces/química , Femenino , Fármacos para la Fertilidad Femenina/administración & dosificación , Fármacos para la Fertilidad Femenina/efectos adversos , Fármacos para la Fertilidad Femenina/farmacología , Histerectomía/efectos adversos , Histerectomía/veterinaria , Células Lúteas/citología , Células Lúteas/metabolismo , Luteinización/efectos de los fármacos , Ovariectomía/efectos adversos , Ovariectomía/veterinaria , Ovario/citología , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovulación/metabolismo , Inducción de la Ovulación/efectos adversos , Inducción de la Ovulación/métodos , Progestinas/administración & dosificación , Distribución Aleatoria , Esteroides/análisis
19.
Biol Reprod ; 87(3): 55, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22674393

RESUMEN

Neovascularization is necessary for formation of the corpus luteum (CL) and includes angiogenesis and vasculogenesis. Vasculogenesis is the formation of new blood vessels by bone marrow-derived endothelial progenitor cells. Here we investigated whether vasculogenesis occurs in neovascularization during CL formation. Mice transplanted with bone marrow from transgenic mice expressing green fluorescent protein (GFP) were injected with equine chorionic gonadotropin and human chorionic gonadotropin (hCG) to induce ovulation and subsequent CL formation. Immunohistochemistry was performed on the ovaries obtained before hCG injection and at 6, 12, and 24 h after hCG injection using antibodies for CD34 or CD31 (an endothelial cell marker), platelet-derived growth factor receptor beta (PDGFR-beta, a pericyte marker), F4/80 (a macrophage marker), and GFP (a bone marrow-derived cell marker). Cells immunostained for CD34, PDGFR-beta, F4/80, and GFP were present in the theca cell layer of the preovulatory follicle before hCG injection. Each of these cell types invaded the granulosa cell layer after hCG injection, and a number of them were observed in the CL 24 h after hCG injection. Fluorescence-based immunohistochemistry or double immunohistochemical staining revealed that a few CD34/CD31-positive cells and PDGFR-beta-positive cells were also positive for GFP in the preovulatory follicle and CL, and that many of the GFP-positive cells recruited to the CL during CL formation were F4/80-positive macrophages. In conclusion, bone marrow-derived vascular progenitor cells and macrophages contribute to neovascularization during CL formation.


Asunto(s)
Células de la Médula Ósea/fisiología , Endotelio Vascular/fisiología , Células Madre Hematopoyéticas/fisiología , Luteinización/fisiología , Neovascularización Fisiológica , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Gonadotropina Coriónica/farmacología , Cuerpo Lúteo/efectos de los fármacos , Cuerpo Lúteo/metabolismo , Cuerpo Lúteo/fisiología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Luteinización/efectos de los fármacos , Luteinización/genética , Luteinización/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Neovascularización Fisiológica/fisiología
20.
J Reprod Dev ; 58(2): 254-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22277932

RESUMEN

The Ski protein is implicated in the proliferation/differentiation of a variety of cells. We previously reported that the Ski protein is present in granulosa cells of atretic follicles, but not in preovulatory follicles, suggesting that Ski has a role in apoptosis of granulosa cells. However, granulosa cells cannot only undergo apoptosis but can alternatively differentiate into luteal cells. It is unknown whether Ski is expressed and has a role in granulosa cells undergoing luteinization. Thus, the aim of the present study was to determine the localization of the Ski protein in the rat ovary during luteinization to examine if Ski might play a role in this process. In order to examine the Ski protein expression during the progression of luteinization, follicular growth was induced in immature female rats by administration of equine chorionic gonadotropin, and luteinization was induced by human chorionic gonadotropin treatment to mimic the luteinizing hormone (LH) surge. While no Ski-positive granulosa cells were present in the preovulatory follicle, Ski protein expression was induced in response to the LH surge and was maintained after formation of the corpus luteum (CL). Although the Ski protein is absent from the granulosa cells of the preovulatory follicle, its mRNA (c-ski) was expressed, and the level of c-ski mRNA was unchanged even after the LH surge. The combined results demonstrated that Ski protein expression is induced in granulosa cells upon luteinization, and suggested that its expression is regulated posttranscriptionally.


Asunto(s)
Cuerpo Lúteo/metabolismo , Células de la Granulosa/metabolismo , Luteinización/metabolismo , Folículo Ovárico/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Procesamiento Postranscripcional del ARN , Animales , Apoptosis/efectos de los fármacos , Gonadotropina Coriónica/farmacología , Cuerpo Lúteo/citología , Cuerpo Lúteo/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Gonadotropinas Equinas/farmacología , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Hipofisectomía , Inmunohistoquímica , Luteinización/efectos de los fármacos , Folículo Ovárico/citología , Folículo Ovárico/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Maduración Sexual , Factores de Tiempo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda