Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Folia Morphol (Warsz) ; 77(2): 301-309, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29064548

RESUMEN

BACKGROUND: This study aimed to investigate the expression and localisation of fol-licle stimulating hormone receptor/growth hormone receptor/luteinising hormone receptor (FSHR/GHR/LHR) in different tissues and examine the regulatory effects of FSHR/GHR/LHR in the reproductive organs of female yaks during luteal phase. MATERIALS AND METHODS: The quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry assays were utilised to analyse the expression and localisation of FSHR/GHR/LHR in different tissues on female yaks. RESULTS: The qRT-PCR results showed that the mRNA expressions of FSHR/GHR/ /LHR were significantly different in the non-reproductive organs (p < 0.01); the highest expression level was observed in the kidney, cerebellum and lung, whereas the lower expression level was observed in the liver and spleen. Im-munohistochemistry assay results showed that FSHR/GHR/LHR were located in kidney tubules, Purkinje cells, cerebellar medulla, alveolar cells and hepato-cytes. In addition, the expression levels of FSHR and GHR were considerably higher than LHR in the reproductive organs of female yaks during luteal phase (p < 0.01). FSHR/GHR/LHR were located in cardiac muscle cells, cerebellar medulla, and theca cell lining of reproductive organs. Furthermore, the expression level of FSHR was higher than those of GHR and LHR in all examined tissues. CONCLUSIONS: Therefore, the expression and localisation of FSHR/GHR/LHR possibly helped to evaluate the effects of them in tissue specific expression on female yaks, investigate the function and mechanism of FSHR/GHR/LHR in the reproductive organs of female yaks during luteal phase. (Folia Morphol 2018; 77, 2: 301-309).


Asunto(s)
Ciclo Estral/fisiología , Regulación de la Expresión Génica/fisiología , Genitales Femeninos/metabolismo , Receptores de HFE/biosíntesis , Receptores de HL/biosíntesis , Receptores de Somatotropina/biosíntesis , Animales , Bovinos , Femenino , Especificidad de Órganos
2.
Mol Reprod Dev ; 84(6): 460-467, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28337831

RESUMEN

Follicle-stimulating hormone receptor (FSHR) is a pivotal regulator of ovarian response to hormonal stimulation. Inflammatory conditions have been linked to lower FSHR expression in granulosa cells (GCs) as well as an attenuated response to hormonal stimulation. The current study aimed to reveal if deficiency and/or blockage of the pro-inflammatory cytokine interleukin 1-alpha (IL1A) increased Fshr expression in rodent GCs. We found elevated Fshr transcript abundance, as assessed by quantitative PCR, in primary GCs isolated from Il1a-knockout compared to wild-type mice, and that the expression of FSHR is significantly higher in Il1a-knockout compared to wild-type ovaries. Supplementing GC cultures with recombinant IL1A significantly lowered Fshr expression in these cells. In accordance with the Fshr expression pattern, proliferation of GCs was higher in follicles from Il1a-knockout mice compared to wild-type mice, as indicated by the MKI67 immunohistochemical staining. Furthermore, treating wild-type mice with anakinra, an IL1 receptor 1 antagonist, significantly increased the expression of Fshr in primary GCs from treated compared to control mice. These data highlight an important interdependency between the potent pro-inflammatory cytokine IL1A and Fshr expression.


Asunto(s)
Regulación de la Expresión Génica , Células de la Granulosa/metabolismo , Interleucina-1alfa/metabolismo , Receptores de HFE/biosíntesis , Animales , Femenino , Células de la Granulosa/citología , Interleucina-1alfa/genética , Ratones , Ratones Noqueados , Receptores de HFE/genética
3.
Immunopharmacol Immunotoxicol ; 38(3): 197-204, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27075695

RESUMEN

CONTEXT: GnRH immunity can reduce the expression of pituitary GnRH levels, and cause the changes in reproductive behaviors. It is unclear whether triptorelin (TRI) and cetrorelix (CET) immunity influences uterine development and expression of follicle-stimulating hormone receptor (FSHR), luteinizing hormone receptor (LHR), and estradiol receptor 1 (ERS1) in the uterus. OBJECTIVE: The study investigated the effects of active immunity of GnRH agonist and antagonist on uterine development, microstructures, expression of hormone receptors mRNAs, and proteins in uteri. MATERIALS AND METHODS: One hundred and five mice were assigned into CET, TRI, and control groups (CG). Mice in CET-1, CET-2, and CET-3 (n = 15) were subcutaneously injected with 10, 20, and 40 µg CET antigens for seven days, respectively. Mice in TRI-1, TRI-2, and TRI-3 were injected with 10, 20, and 40 µg TRI antigens for seven days, respectively. The qPCR and Western blot were implemented to determine expressions of ESR1, LHR and FSHR mRNAs, and proteins. RESULTS: Compared with CG, the uterine weights of CET-1, CET-2, and CET-3 increased by 42.86, 62.86, and 10.00% on day 35 (p < 0.05), respectively. Uterine weights of TRI-2, TRI-3 reduced by 28.57% and 11.43% (p < 0.05), respectively. The uterine cavity in CET-1, CET-2, and CET-3 increased; the uterine wall became thick. The cytoplasm of endometrial epithelial cells (EEC) increased slightly. In TRI group, the uterine wall thinned. Uterine cavity became narrow slightly in TRI-1. Numbers of uterine glands reduced. The endometrium epithelial thickness (EET) in CET-1 and CET-2 increased by 68.21% and 79.46% (p < 0.05), respectively. EET in TRI-1 was decreased by 13.69%. Uterine wall thicknesses (UWT) in CET-1 and CET-2 were higher than CG, with the increment of 28.59% and 30.72%. UWT of TRI-1, TRI-2, and TRI-3 reduced by 29.35, 15.36, and 14.41%, respectively. Expressions of ESR1, FSHR, and LHR mRNAs in CET and TRI mice increased. ESR1 and FSHR protein levels increased in all experimental mice (p < 0.05), with a maximum of TRI-3. LHR protein levels of the CET decreased. LHR protein levels of TRI group increased, with a maximum of TRI-3 (p < 0.05). ESR1 protein level had significant negative correlations to mRNA expressions of ESR1, LHR, and FSHR. CONCLUSIONS: CET immunity promoted the uterine development, improved EET and UWT, and also promoted the expressions of ESR1 and FSHR protein levels. It lessened the LHR protein levels. TRI immunity blocked EET and UWT, inhibited uterine growth and development. The efficacy of CET immunity was more obvious than TRI.


Asunto(s)
Receptor alfa de Estrógeno/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/análogos & derivados , Receptores de HFE/biosíntesis , Receptores de HL/biosíntesis , Pamoato de Triptorelina/farmacología , Útero/crecimiento & desarrollo , Animales , Receptor alfa de Estrógeno/inmunología , Femenino , Regulación de la Expresión Génica/inmunología , Hormona Liberadora de Gonadotropina/farmacología , Ratones , Receptores de HFE/inmunología , Receptores de HL/inmunología , Útero/inmunología
4.
J Biol Regul Homeost Agents ; 28(3): 419-31, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25316123

RESUMEN

The process of granulosa cell luteinization is part of the main process determining growth, differentiation and proliferation of these cells. Although the mechanisms underlying the regulation of luteinizing hormone receptor (LHR), follicle stimulating hormone receptor (FSHR) and cytochrome P450 aromatase expression in mammalian granulosa cells is well understood, still little is known about the expression of mRNA and encoded proteins in relation to cell proliferation and luteinization in vitro. Porcine granulosa cells were observed in vitro at a168-h period while undergoing real-time proliferation using an RTCA system. Furthermore, LHR, FSHR and CYP19 mRNA expression were detected using RQ-PCR after 168 h of in vitro culture (IVC) at 24-h intervals, and LHR, FSHR and P450arom were examined by confocal microscopic observation at 0 h, 24 h, 48 h, 96 h, and 168 h of IVC. We found increased expression of LHR and CYP19 mRNA at 24 h and 48 h of IVC compared to the other stages (P less than 0.01, P less than 0.001), whereas FSHR mRNA was higher only at 0 h (P less than 0.001). In contrast, LHR, FSHR and P450arom protein expression was significantly higher at the end of the 168-h IVC period compared to 0 h, 24 h, 48 h and 96 h (P less than 0.001). LHR, FSHR and P450arom were distributed in the cytoplasm of porcine GCs at each time point of IVC. When analyzing cell proliferation, differences in cell index were observed (at least P less than 0.05) between the first (0-24 h) and the last period (144-168 h) of IVC; however, soon after 24 h of IVC a logarithmic increase in proliferation was also seen. We assume that the expression of LHR, FSHR and CYP19 mRNAs depends on the period of in vitro cultivation and may be linked with the luteinization process of porcine GCs. Furthermore, the patterns of mRNA and protein expression suggest a post-transcriptional regulation of LHR, FSHR and P450arom. In summary, it can be presumed that mRNA and protein expression and in vitro luteinization and proliferation of porcine GCs are regulated by different mechanisms, because not all of these processes are correlated.


Asunto(s)
Aromatasa/biosíntesis , Proliferación Celular , Regulación de la Expresión Génica/fisiología , Células de la Granulosa/metabolismo , Receptores de HFE/biosíntesis , Receptores de HL/biosíntesis , Animales , Femenino , Células de la Granulosa/citología , ARN Mensajero/biosíntesis , Porcinos
5.
Mol Hum Reprod ; 19(8): 519-27, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23562944

RESUMEN

Anti-Müllerian hormone (AMH) is exclusively produced by granulosa cells (GC) of the developing pre-antral and antral follicles, and AMH is increasingly used to assess ovarian function. It is unclear which size follicles make the most AMH (total content) and are the main contributors to circulating AMH concentrations. To determine AMH gene expression in GC (q-RT-PCR) and follicular AMH production (Elisa and RIA) in relation to follicular development, 87 follicles (3-13 mm diameter) including both GC and the corresponding follicular fluid (FF) were collected in connection with fertility preservation of human ovaries. Further, follicle number and diameter, graded in 1 mm increments, were determined by 3D ultrasound in 113 women in their natural menstrual cycle to determine follicle number and diameter in relation to circulating AMH levels. This study demonstrates for the first time a positive association between AMH gene expression in human and both total follicular fluid AMH (P < 0.02) and follicular fluid AMH concentration (P < 0.01). AMH gene expression and total AMH protein increased until a follicular diameter of 8 mm, after which a sharp decline occurred. In vivo modelling confirmed that 5-8 mm follicles make the greatest contribution to serum AMH, estimated for the first time in human to be 60% of the circulating concentration. Significant positive associations between gene expression of AMH and FSHR, AR and AMHR2 expression (P < 0.00001 for all three) and significant negative association between follicular fluid AMH concentration and CYP19a1 expression were found (P < 0.0001). Both AMH gene expression (P < 0.02) and follicular fluid concentration of AMH (P < 0.00001) correlated negatively with estradiol concentration.


Asunto(s)
Hormona Antimülleriana/biosíntesis , Hormona Antimülleriana/metabolismo , Líquido Folicular/metabolismo , Células de la Granulosa/metabolismo , Adolescente , Adulto , Hormona Antimülleriana/genética , Aromatasa/biosíntesis , Niño , Estradiol/sangre , Femenino , Expresión Génica , Humanos , Receptores de HFE/biosíntesis , Receptores de Péptidos/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Adulto Joven
6.
Reprod Fertil Dev ; 25(8): 1194-203, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23241220

RESUMEN

The present study investigated the role of growth differentiation factor (GDF)-9 and FSH, alone or in combination, on the growth, viability and mRNA expression of FSH receptor, proliferating cell nuclear antigen (PCNA) and proteoglycan-related factors (i.e., hyaluronan synthase (HAS) 1, HAS2, versican, perlecan) in bovine secondary follicles before and after in vitro culture. After 12 days culture, sequential FSH (100 ng mL⁻¹) from Days 0 to 6 and 500 ng mL⁻¹ from Days 7 to 12) increased follicular diameter and resulted in increased antrum formation (P<0.05). Alone, 200 ng mL⁻¹ GDF-9 significantly reduced HAS1 mRNA levels, but increased versican and perlecan mRNA levels in whole follicles, which included the oocyte, theca and granulosa cells. Together, FSH and GDF-9 increased HAS2 and versican (VCAN) mRNA levels, but decreased PCNA mRNA expression, compared with levels in follicles cultured in α-minimum essential medium supplemented with 3.0 mg mL⁻¹ bovine serum albumin, 10 µg mL⁻¹ insulin, 5.5 µg mL⁻¹ transferrin, 5 ng mL⁻¹ selenium, 2 mM glutamine, 2mM hypoxanthine and 50 µg mL⁻¹ ascorbic acid (α-MEM⁺). Comparisons of uncultured (0.2 mm) and α-MEM⁺ cultured follicles revealed that HAS1 mRNA expression was higher, whereas VCAN expression was lower, in cultured follicles (P<0.05). Expression of HAS1, VCAN and perlecan (HSPG2) was higher in cultured than in vivo-grown (0.3 mm) follicles. In conclusion, FSH and/or GDF-9 promote follicular growth and antrum formation. Moreover, GDF-9 stimulates expression of versican and perlecan and interacts positively with FSH to increase HAS2 expression.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factor 9 de Diferenciación de Crecimiento/metabolismo , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Oogénesis , Folículo Ovárico/metabolismo , ARN Mensajero/metabolismo , Mataderos , Animales , Bovinos , Supervivencia Celular , Femenino , Líquido Folicular/enzimología , Líquido Folicular/metabolismo , Glucuronosiltransferasa/antagonistas & inhibidores , Glucuronosiltransferasa/biosíntesis , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Hialuronano Sintasas , Isoenzimas/antagonistas & inhibidores , Isoenzimas/biosíntesis , Isoenzimas/genética , Isoenzimas/metabolismo , Oocitos/citología , Oocitos/enzimología , Oocitos/metabolismo , Folículo Ovárico/citología , Folículo Ovárico/crecimiento & desarrollo , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Antígeno Nuclear de Célula en Proliferación/química , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/biosíntesis , Proteoglicanos/genética , Proteoglicanos/metabolismo , Receptores de HFE/antagonistas & inhibidores , Receptores de HFE/biosíntesis , Receptores de HFE/genética , Receptores de HFE/metabolismo , Técnicas de Cultivo de Tejidos/veterinaria
7.
Gen Comp Endocrinol ; 185: 57-66, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23395683

RESUMEN

To determine whether hormone-receptor signaling pathways in the thymus are altered by active immunization against gonadotrophin-releasing hormone I (GnRH), 3-week-old Sprague-Dawley male rats received GnRH-tandem-OVA peptides (200 µg/ml), and the effects were compared to a control group. Serum testosterone, LH and FSH concentrations were markedly reduced, with severe testicular atrophy, compared to controls, demonstrating effective blockade of the pituitary-gonadal axis. The reduction in LH and FSH concentrations in the thymus of immunized animals was lower than that observed in the serum, where a significant difference (P<0.001) in concentration was observed between both groups. Concentrations of GnRH were increased in the thymus of immunized rats. In thymic tissue, GnRHR, FSHR and LHR demonstrated stronger immunostaining, and AR weaker staining, in the immunized group compared to controls. Reproductive hormone receptor mRNA expression was consistent with protein variations in the immunized thymus. Compared to controls, GnRHR gene levels were significantly increased (P<0.05), however, AR mRNA expression were greatly decreased with immune week-age (P<0.05). Both FSHR and LHR mRNA expression levels were significantly higher in the treated group than in controls in the first three samples (P<0.05). When GnRHR was blocked by an antagonist in thymocytes, all reproductive hormone receptor gene expressions were significantly increased (P<0.001). In summary, these findings suggest that active immunization against GnRH can up-regulate GnRH receptor and gonadotropin receptor signaling, by stimulating thymic autocrine and paracrine function, whereas the androgen receptor is down-regulated due to a lack of testosterone secretion in the thymus.


Asunto(s)
Hormona Liberadora de Gonadotropina/inmunología , Receptores de HFE/biosíntesis , Receptores LHRH/biosíntesis , Receptores de HL/biosíntesis , Timo/metabolismo , Animales , Hormona Folículo Estimulante/sangre , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Luteinizante/sangre , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de HFE/genética , Receptores de HL/genética , Receptores LHRH/genética , Testículo/inmunología , Testosterona/sangre , Timo/inmunología , Vacunación
8.
Biochem Biophys Res Commun ; 422(1): 54-8, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22561017

RESUMEN

A direct effect of FSH on bone turnover via stimulation of osteoclast formation has been reported. Here we show that monoclonal or polyclonal antibodies to FSH inhibit osteoclast formation induced by FSH to an extent similar to that noted in FSH receptor (FSHR) knockout cells. Furthermore, we document the amplification of FSHR cDNA from well-characterized human CD14+ osteoclast precursors and osteoclasts, and the direct sequencing of the PCR products to definitively establish the expression of FSHRs. At these sites, the FSHR was expressed predominantly as an isoform that omits exon 9, a linker between the FSH-binding region and a long, invariant signaling domain of the receptor. These data provide compelling evidence for expression of a FSH receptor isoform in osteoclasts and their precursors.


Asunto(s)
Resorción Ósea/metabolismo , Hormona Folículo Estimulante de Subunidad beta/fisiología , Osteoclastos/fisiología , Receptores de HFE/biosíntesis , Empalme Alternativo , Animales , Anticuerpos Monoclonales , Secuencia de Bases , Células Cultivadas , Femenino , Hormona Folículo Estimulante de Subunidad beta/antagonistas & inhibidores , Humanos , Ratones , Datos de Secuencia Molecular , Osteoclastos/efectos de los fármacos , Ovario , Receptores de HFE/genética , Transcripción Genética
9.
Dev Biol ; 334(2): 458-67, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19666016

RESUMEN

Targeted disruption of the inhibin alpha gene (Inha(-)(/)(-)) in mice results in an ovarian phenotype of granulosa cell tumors that renders the animals infertile. Little is known about the reproductive defects prior to tumor development. Here, we report novel data on early follicle dynamics in Inha(-)(/)(-) mice, which demonstrate that inhibin alpha has important consequences upon follicle development. Morphological changes in both germ and somatic cells were evident in postnatal day 12 ovaries, with Inha(-/-) mice exhibiting numerous multilayered follicles that were far more advanced than those observed in age-matched controls. These changes were accompanied by alterations in follicle dynamics such that Inha(-/-) ovaries had fewer follicles in the resting pool and more committed in the growth phase. Absence of inhibin alpha resulted in advanced follicular maturation as marked by premature loss of anti-Müllerian hormone (AMH) in secondary follicles. Additionally, gene expression analysis revealed changes in factors known to be vital for oocyte and follicle development. Together, these data provide key evidence to suggest that regulation of the inhibin/activin system is essential for early folliculogenesis in the prepubertal mouse ovary.


Asunto(s)
Tumor de Células de la Granulosa/genética , Células de la Granulosa/patología , Inhibinas/deficiencia , Oocitos/patología , Folículo Ovárico/patología , Neoplasias Ováricas/genética , Factores de Edad , Animales , Aromatasa/biosíntesis , Aromatasa/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Femenino , Hormona Folículo Estimulante/sangre , Regulación de la Expresión Génica , Tumor de Células de la Granulosa/patología , Inhibinas/genética , Inhibinas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Folículo Ovárico/fisiopatología , Neoplasias Ováricas/patología , Receptores de HFE/biosíntesis , Receptores de HFE/genética , Maduración Sexual
10.
Reproduction ; 140(4): 551-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20639315

RESUMEN

Prior to follicle selection into the preovulatory hierarchy, hen granulosa cells from prehierarchal follicles remain undifferentiated, as defined in part by the virtual absence of LHR mRNA expression and inability to produce progesterone. It has previously been proposed that prior to follicle selection, granulosa cells are actively maintained in an undifferentiated state by epidermal growth factor receptor ligands (EGFRL) signaling via the MAP kinase/extracellular regulated kinase pathway. Moreover, there is recent evidence that EGFRL/MAP kinase signaling modulates FSH receptor (FSHR) transcription, in part, via inhibitor of differentiation/DNA-binding (ID) proteins. In the present studies with undifferentiated granulosa, recombinant human (rh) bone morphogenetic protein 2 (BMP2) induced the phosphorylation of SMAD1/5/8, and blocked transforming growth factor ß and FSH-induced FSHR expression and progesterone production. Significantly, BMP2 rapidly induced mRNAs encoding betacellulin and EGF, plus ID proteins (ID1, ID3, and ID4). Alternatively, the bioactivity of BMPs can be modulated by one or more BMP antagonists, including noggin (NOG). NOG mRNA is expressed by both hen granulosa and theca tissues from prehierarchal follicles. Pretreatment of cultured granulosa with rh NOG reversed both the stimulatory effects of BMP2 on ID1, ID3, and ID4 expression and the inhibitory effects of BMP2 on FSHR mRNA levels and progesterone production. Collectively, these data provide evidence that prior to follicle selection, BMP2 signaling contributes toward maintaining granulosa cells in an undifferentiated state. The actions of BMP2 are, at least in part, mediated indirectly via enhanced EGFRL expression and ERBB receptor-mediated MAP kinase signaling, and can be modulated by the autocrine/paracrine production of NOG.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Pollos/metabolismo , Hormona Folículo Estimulante/antagonistas & inhibidores , Células de la Granulosa/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Folículo Ovárico/metabolismo , Receptores de HFE/metabolismo , Animales , Western Blotting/veterinaria , Proteína Morfogenética Ósea 2/biosíntesis , Proteína Morfogenética Ósea 2/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diferenciación Celular/fisiología , Femenino , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/citología , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Folículo Ovárico/citología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Ribosómico 18S/química , ARN Ribosómico 18S/genética , Receptores de HFE/biosíntesis , Receptores de HFE/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria
11.
Microsc Res Tech ; 82(6): 635-642, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30582244

RESUMEN

Many studies have reported that human endometrial mesenchymal stem cells (HuMenSCs) are capable of repairing damaged tissues. The aim of the present study was to investigate the effects of HuMenSCs transplantation as a treatment modality in premature ovarian failure (POF) associated with chemotherapy-induced ovarian damage. HuMenSCs were isolated from menstrual blood samples of five women. After the in vitro culture of HuMenSCs, purity of the cells was assessed by cytometry using CD44, CD90, CD34, and CD45 FITC conjugate antibody. Twenty-four female Wistar rats were randomly divided into four groups: negative control, positive control, sham, and treatment groups. The rat models of POF used in our study were established by injecting busulfan intraperitoneally into the rats during the first estrus cycle. HuMenSCs were transplanted by injection via the tail vein into the POF-induced rats. Four weeks after POF induction, ovaries were collected and the levels of Amh, Fst, and Fshr expression in the granulosa cell (GC) layer, as well as plasma estradiol (E2) and progesterone (P4) levels were evaluated. Moreover, migration and localization of DiI-labeled HuMenSCs were detected, and the labeled cells were found to be localized in GCs layer of immature follicles. In addition to DiI-labelled HuMenSCs tracking, increased levels of expression of Amh and Fshr and Fst, and the high plasma levels of E2 and P4 confirmed that HuMenSC transplantation had a significant effect on follicle formation and ovulation in the treatment group compared with the negative control (POF) group.


Asunto(s)
Trasplante de Células/métodos , Células de la Granulosa/fisiología , Células Madre Mesenquimatosas/fisiología , Insuficiencia Ovárica Primaria/terapia , Animales , Hormona Antimülleriana/biosíntesis , Busulfano/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Hormona Folículo Estimulante/biosíntesis , Perfilación de la Expresión Génica , Histocitoquímica , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Folículo Ovárico/patología , Ovario/patología , Ovario/fisiología , Ovulación , Insuficiencia Ovárica Primaria/inducido químicamente , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de HFE/biosíntesis , Resultado del Tratamiento
12.
Oncol Res ; 17(5): 231-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18980020

RESUMEN

Epithelial ovarian cancer (EOC) has remained an enigmatic disease, the etiology of which is mostly unknown. Considering the age incidence around menopause, a "gonadotrophin theory" has been proposed, and considering the association with infertility, an "incessant ovulation" theory has been proposed. EOC originates from ovarian surface epithelium (OSE), which not only secretes cytokines/growth factors and steroids but expresses gonadotrophin and steroid hormone receptors as well. The most important gonadotrophin receptor is the follicle stimulating hormone receptor (FSHR), which has definite oncogenic potential and is a probable candidate for oncogenesis. In this article, we review existing knowledge of FSHR in ovary, in OSE, and in epithelial ovarian cancer and try to establish relative importance of this receptor over its ligand. A systematic review through PubMed was done on the subject of FSHR and its metabolism. For the obvious difficulty of meager amounts of tissue available, most of studies so far see it in granulosa cells and cell lines rather than in OSE. Thus, effort was made to deduce workable knowledge that can establish its role and can then be applied in OSE and EOC. There is great deal of information regarding metabolism of FSHR, including regulation of its gene expression, isoforms, and pathways of desensitization and degradation with which ovarian cancer etiology researchers have to be familiar with, and there are a number of steps where manipulation may stop carcinogenesis. Hormone therapy of such cancer has so far been only mildly active, probably because we do not understand the role and mechanism of action of FSH and FSHR in the hypothalamo-pituitary-ovarian axis in development of such cancer.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Neoplasias Ováricas/metabolismo , Ovario/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Hormona Folículo Estimulante/biosíntesis , Hormona Folículo Estimulante/genética , Humanos , Neoplasias Ováricas/genética , Receptores de HFE/biosíntesis , Receptores de HFE/genética , Receptores de HFE/metabolismo
13.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 28(3): 242-4, 2008 Mar.
Artículo en Zh | MEDLINE | ID: mdl-18476426

RESUMEN

OBJECTIVE: To explore the regulatory effect and mechanism of Ningxin Hongqi Capsule on local ovarian autocrine and paracrine factors in peri-menopausal rats. METHODS: SD female rats aged 4 months were allocated in a normal control group (A) and those aged 14 months with vagino-cytologic figure of oestrus elongation were allocated in a senile female rat model group (B). Rats in Group B were subdivided into 5 groups randomly as the B1, B2 and B3 subgroups treated respectively with high, moderate and low dose Ningxin Hongqi Capsule, the B4 subgroup treated with estradiol and the B5 subgroup untreated for control. Rats' ovaries were obtained at the end of the experiment for observing the conditions of ovarian growing follicles and corpus luteum by HE staining, determining expressions of ovarian estradiol receptor (ER), progesterone receptor (PR), follicle-stimulating hormone (FSH), luteinizing hormone (LH), inhibin alpha (INHalpha), activin (ACT) alpha-beta, follistatin (FS), and insulin-like growth factor (IGF-1). RESULTS: As compared with Group B5, the ovary index, number of growing follicle were higher and levels of FSH and LH were lower in Group B2 and B3, expression of ER was higher in Group B1 and B4, IGF-1 and INHalpha was higher in Group B2 and B3, and ACTalpha-beta and FS were lower (all P < 0.05). CONCLUSION: Nirigxin Hongqi Capsule could adjust and balance the local ovarian autocrine and paracrine factors to improve the ovarian function.


Asunto(s)
Comunicación Autocrina/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Ovario/efectos de los fármacos , Comunicación Paracrina/efectos de los fármacos , Animales , Comunicación Autocrina/fisiología , Cápsulas , Femenino , Humanos , Modelos Animales , Ovario/metabolismo , Ovario/fisiología , Comunicación Paracrina/fisiología , Perimenopausia , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores de Estradiol/biosíntesis , Receptores de HFE/biosíntesis , Receptores de Progesterona/biosíntesis
14.
J Clin Invest ; 102(7): 1352-9, 1998 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9769327

RESUMEN

A single natural loss of function mutation of the follicle stimulating hormone receptor (FSHR) has been described to date. Present in the Finnish population it markedly impairs receptor function, blocking follicle development at the primary stage and presenting as primary amenorrhea with atrophic ovaries. When Western European women with this phenotype were examined for FSHR mutations the result was negative, suggesting that other etiologies corresponding to this clinical pattern are markedly more frequent. We now describe a novel phenotype related to mutations provoking a partial loss of function of the FSHR. A woman with secondary amenorrhea had very high plasma gonadotropin concentrations (especially FSH), contrasting with normal sized ovaries and antral follicles up to 5 mm at ultrasonography. Histological and immunohistochemical examination of the ovaries showed normal follicular development up to the small antral stage and a disruption at further stages. The patient was found to carry compound heterozygotic mutations of the FSHR gene: Ile160Thr and Arg573Cys substitutions located, respectively, in the extracellular domain and in the third intracellular loop of the receptor. The mutated receptors, when expressed in COS-7 cells, showed partial functional impairment, consistent with the clinical and histological observations: the first mutation impaired cell surface expression and the second altered signal transduction of the receptor. This observation suggests that a limited FSH effect is sufficient to promote follicular growth up to the small antral stage. Further development necessitates strong FSH stimulation. The contrast between very high FSH levels and normal sized ovaries with antral follicles may thus be characteristic of such patients.


Asunto(s)
Amenorrea/genética , Infertilidad Femenina/genética , Mutación Puntual , Receptores de HFE/genética , Adulto , Amenorrea/sangre , Amenorrea/diagnóstico por imagen , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Células COS , Bovinos , Membrana Celular/fisiología , Europa (Continente) , Femenino , Finlandia , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante/metabolismo , Heterocigoto , Humanos , Infertilidad Femenina/sangre , Cinética , Masculino , Ratones , Modelos Moleculares , Ovario/diagnóstico por imagen , Ovario/patología , Linaje , Fenotipo , Conformación Proteica , Ratas , Receptores de HFE/biosíntesis , Receptores de HFE/fisiología , Proteínas Recombinantes/biosíntesis , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Ovinos , Transducción de Señal , Porcinos , Transfección , Ultrasonografía
15.
Exp Biol Med (Maywood) ; 232(8): 1050-63, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17720951

RESUMEN

The hypothalamic-pituitary-gonadal (HPG) axis and the somatotropic axis are influenced by nutritional factors. Calorie restriction (CR) extends lifespan but suppresses both the HPG and the somatotropic axes. Since most CR studies use a fairly severe (40%-60%) reduction of calorie intake, we hypothesized that a milder CR (20%) might not be deleterious to reproduction in male mice. To test this hypothesis, we evaluated the effects of 20% CR on testicular testosterone content and on testicular expression of genes that are relevant to testicular function and reproductive competence, including insulin-like growth factor-I, cytochrome P450 aromatase (Cyp19a1), androgen receptor, luteinizing hormone receptor, follicle-stimulating hormone receptor, cytochrome P450c17 and 3-beta-hydroxysteroid dehydrogenase/isomerase. To relate CR effects to the activity of the somatotropic axis, we have used growth hormone-resistant GHR knockout mice as well as transgenic mice overexpressing GH. Mild CR did not affect testosterone levels in testis homogenates and had little effect on expression of the examined genes in the reproductive organs. Altered activity of the GH/insulin-like growth factor-1 axis had a major impact on the parameters analyzed. The results also suggest that expression of several key genes involved in the control of testicular function is preserved under conditions of mild CR and encourage speculation that mild regimens of CR can produce longevity benefits without impairing reproduction.


Asunto(s)
Restricción Calórica , Regulación de la Expresión Génica/fisiología , Longevidad/fisiología , Reproducción/fisiología , Testículo/metabolismo , Testosterona/biosíntesis , 3-Hidroxiesteroide Deshidrogenasas/biosíntesis , Animales , Aromatasa/biosíntesis , Hormona del Crecimiento/genética , Hormona del Crecimiento/metabolismo , Sistema Hipotálamo-Hipofisario/fisiología , Factor I del Crecimiento Similar a la Insulina/biosíntesis , Masculino , Ratones , Ratones Noqueados , Receptores Androgénicos/biosíntesis , Receptores de HFE/biosíntesis , Receptores de Somatotropina/deficiencia , Receptores de Somatotropina/metabolismo
16.
Biosci Rep ; 27(6): 403-11, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17968653

RESUMEN

Sertoli cells are regulated by follicular stimulating hormone (FSH) and testosterone secreted by the pituitary gland and Leydig cells, respectively. However, the expression of the FSH receptor and androgen receptor were undetectable in both primary cultured Sertoli cells and Sertoli cell lines immortalized by SV40 large T antigen. Two Sertoli cell lines, B6Sc-2 and B6Sc-3, were established from the testis of 19-day-old C57BL/6 mice testis by immortalization with human telomere reverse transcriptase. These Sertoli cell lines expressed FSH receptors and the total phosphoprotein patterns were converted after FSH treatment. Additionally, immunological methods demonstrated that these cell lines expressed characteristic Sertoli cell proteins, such as tyrosine-tubulin, vimentin and stem cell factor (SCF). Reverse transcription-polymerase chain reaction (RT-PCR) also indicates that they express Sertoli specific mRNAs, such as Amh, claudin11 and ZO-1. The expression of the androgen receptor in both B6Sc-2 and B6Sc-3 cells could be induced by TNF-alpha treatment. The present results indicate that these Sertoli cell lines are more native than others and may thus provide useful tools for in vitro studies.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Receptores Androgénicos/biosíntesis , Células de Sertoli/metabolismo , Telomerasa/biosíntesis , Factor de Necrosis Tumoral alfa/farmacología , Animales , Antígenos Virales de Tumores/biosíntesis , Antígenos Virales de Tumores/genética , Línea Celular Transformada , Hormona Folículo Estimulante/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Especificidad de Órganos/efectos de los fármacos , Especificidad de Órganos/genética , Hipófisis/metabolismo , Receptores Androgénicos/genética , Receptores de HFE/biosíntesis , Receptores de HFE/genética , Células de Sertoli/citología , Telomerasa/genética , Testosterona/metabolismo
17.
J Ovarian Res ; 10(1): 32, 2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28472976

RESUMEN

BACKGROUND: Diminished ovarian reserve(DOR) is associated with female infertility and poor response to ovarian stimulation. Our objective was to assess the effect of dehydroepiandrosterone(DHEA) on DOR women and to explore whether the improvement of ovarian response after DHEA supplementation was dependent on the expression levels of androgen receptor(AR). METHODS: A prospective cohort study was performed in the Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital during August 2014 to August 2016. 103 DOR women who completed the study were divided into the DHEA group (n = 53), which received DHEA supplementation (25 mg three times a day) for 8 weeks, and the control group (n = 50), which did not receive DHEA, before the IVF cycles. Serum hormone levels(FSH, LH, E2, T, DHEAs, AMH, INHB), antral follicle count(AFC) and the expression of AR and FSH receptor(FSHR) in granulosa cells(GCs) were measured, meanwhile ovarian response parameters and IVF outcomes were compared. The GCs from another 36 DOR women were cultured with different concentrations of DHEA in vitro. Then, we compared the expression of AR and FSHR in GCs according to the different numbers of oocytes retrieved both in DHEA and control group. RESULTS: In the present study, DHEA supplementation resulted in significantly higher levels of serum T(P = 0.047), DHEAs(P = 0.019) and AR mRNA expression in GCs(P = 0.049). In vitro experiment, the protein and mRNA expression of AR and FSHR in the preovulatory GCs were significantly increased in response to DHEA supplementation(P <0.05). No significant differences were found in ovarian reserve, ovarian response, or IVF outcomes between the two groups. Subgroup analyses showed the levels of AR and FSHR mRNA in GCs were significantly increased in DHEA group with ≥5 oocytes retrieved(P <0.05). CONCLUSION: DHEA supplementation can increase the expression of AR in preovulatory GCs both in vivo and in vitro. The selective beneficial effects of DHEA supplementation on ovarian response in DOR women may depend on the increasing expression of AR and FSHR in GCs. TRIAL REGISTRATION: The Chinese Clinical Trial Registry ( ChiCTR-IPR-15006126 ). Retrospectively Registered 19 March 2015.


Asunto(s)
Deshidroepiandrosterona/farmacología , Infertilidad Femenina/terapia , Reserva Ovárica/efectos de los fármacos , Ovario/efectos de los fármacos , Receptores Androgénicos/biosíntesis , Adulto , Deshidroepiandrosterona/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Fertilización In Vitro/métodos , Células de la Granulosa/efectos de los fármacos , Humanos , Infertilidad Femenina/metabolismo , Infertilidad Femenina/fisiopatología , Ovario/metabolismo , Inducción de la Ovulación/métodos , Embarazo , Resultado del Embarazo , Estudios Prospectivos , ARN Mensajero/genética , Receptores Androgénicos/genética , Receptores de HFE/biosíntesis , Receptores de HFE/genética , Regulación hacia Arriba/efectos de los fármacos
18.
Biomed Res Int ; 2017: 9738640, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28337462

RESUMEN

The key mechanisms responsible for achievement of full reproductive and developmental capability in mammals are the differentiation and transformation of granulosa cells (GCs) during folliculogenesis, oogenesis, and oocyte maturation. Although the role of 17 beta-estradiol (E2) in ovarian activity is widely known, its effect on proliferative capacity, gap junction connection (GJC) formation, and GCs-luteal cells transformation requires further research. Therefore, the goal of this study was to assess the real-time proliferative activity of porcine GCs in vitro in relation to connexin (Cx), luteinizing hormone receptor (LHR), follicle stimulating hormone receptor (FSHR), and aromatase (CYP19A1) expression during short-term (168 h) primary culture. The cultured GCs were exposed to acute (at 96 h of culture) and/or prolonged (between 0 and 168 h of culture) administration of 1.8 and 3.6 µM E2. The relative abundance of Cx36, Cx37, Cx40, Cx43, LHR, FSHR, and CYP19A1 mRNA was measured. We conclude that the proliferation capability of GCs in vitro is substantially associated with expression of Cxs, LHR, FSHR, and CYP19A1. Furthermore, the GC-luteal cell transformation in vitro may be significantly accompanied by the proliferative activity of GCs in pigs.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células de la Granulosa/metabolismo , Oogénesis/efectos de los fármacos , Animales , Diferenciación Celular/genética , Familia 19 del Citocromo P450/biosíntesis , Estradiol/administración & dosificación , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Humanos , Técnicas de Maduración In Vitro de los Oocitos , Oocitos/efectos de los fármacos , Oocitos/crecimiento & desarrollo , Oogénesis/genética , Receptores de HFE/biosíntesis , Receptores de HFE/genética , Receptores de HL/biosíntesis , Receptores de HL/genética , Porcinos
19.
J Clin Endocrinol Metab ; 91(6): 2396-403, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16537688

RESUMEN

CONTEXT: Previous studies showed that nerve growth factor (NGF) induces the expression of functional FSH receptors (FSHR) in preantral follicles of the developing rat ovary. OBJECTIVE: The objective of this study was to determine whether NGF can affect granulosa cell (GC) function in human periovulatory follicles using intact human ovaries and isolated human GCs. PATIENTS AND INTERVENTIONS: Human GCs were obtained from in vitro fertilization patients and normal ovaries from women with elective pelvic surgery for nonovarian indications. RESULTS: In normal ovaries, NGF and trkA (NGF's high-affinity receptor) were detected by immunohistochemistry in GCs of preantral and antral follicles. NGF and trkA are also present in thecal cells of antral follicles. Both freshly collected and cultured GCs contained immunoreactive NGF and trkA in addition to their respective mRNAs. Human GCs respond to NGF with increased estradiol (E(2)) secretion and a reduction in progesterone output. Exposure of human GCs to NGF increased FSHR mRNA content within 18 h of treatment, and this effect was blocked by the trk tyrosine kinase blocker K-252a. Also, cells preexposed to NGF released significantly more E(2) in response to hFSH than cells not pretreated with the neurotropin, showing that the NGF-induced increase in FSHR gene expression results in the formation of functional FSHRs. CONCLUSIONS: These results suggest that one of the functions of NGF in the preovulatory human ovary is to increase the secretion of E(2) while preventing early luteinization via an inhibitory effect on progesterone secretion. NGF stimulates E(2) secretion both directly and by increasing the formation of FSHRs.


Asunto(s)
Estradiol/metabolismo , Células de la Granulosa/metabolismo , Factor de Crecimiento Nervioso/farmacología , Receptor trkA/fisiología , Receptores de HFE/biosíntesis , Femenino , Humanos , Progesterona/metabolismo , ARN Mensajero/análisis , Receptores de HFE/genética
20.
Mol Cell Endocrinol ; 247(1-2): 73-81, 2006 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-16406266

RESUMEN

Sertoli cells support the development of germ cells by providing a microenvironment in the seminiferous tubules. FSH stimulates Sertoli cell proliferation only during neonatal period till day 18 in the immature rat whereas FSH regulates only functional parameters in the adult rat Sertoli cells. This suggests that FSH exerts differential action in immature and adult Sertoli cells. In an attempt to elucidate the mechanism by which FSH exerts the differential effects, we have carried out both in vivo and in vitro studies using Sertoli cells isolated from immature (7-10 days old) and adult (90 days old) rats. The differential role of FSH was studied at the receptor as well as at the signaling level. Monitoring the level of expression of FSH receptor by RTPCR and northern blot analysis revealed that the expression was more in immature Sertoli cells. Furthermore, it was found that FSH up (1.8-fold) regulates its receptor level only in the immature Sertoli cells and not in the adult. Results also revealed that PKIbeta and calcium, which are the downstream signaling molecules, are involved in FSH regulated Sertoli cells proliferation. It was also observed that FSH up (1.4-fold) regulates the levels of expression of IL-6 mRNA only in the immature rat Sertoli cells suggesting the possibility of its involvement in FSH regulated Sertoli cell proliferation.


Asunto(s)
Calcio/metabolismo , Hormona Folículo Estimulante/fisiología , Interleucina-6/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Biomarcadores/metabolismo , Proliferación Celular , Células Cultivadas , Masculino , Ratas , Ratas Wistar , Receptores de HFE/biosíntesis , Células de Sertoli/fisiología , Transducción de Señal , Transferrina/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA