Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Microsc Res Tech ; 82(6): 635-642, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30582244

RESUMO

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.


Assuntos
Transplante de Células/métodos , Células da Granulosa/fisiologia , Células-Tronco Mesenquimais/fisiologia , Insuficiência Ovariana Primária/terapia , Animais , Hormônio Antimülleriano/biossíntese , Bussulfano/administração & dosagem , Modelos Animais de Doenças , Feminino , Hormônio Foliculoestimulante/biossíntese , Perfilação da Expressão Gênica , Histocitoquímica , Humanos , Injeções Intraperitoneais , Injeções Intravenosas , Folículo Ovariano/patologia , Ovário/patologia , Ovário/fisiologia , Ovulação , Insuficiência Ovariana Primária/induzido quimicamente , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Receptores do FSH/biossíntese , Resultado do Tratamento
2.
Folia Morphol (Warsz) ; 77(2): 301-309, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29064548

RESUMO

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).


Assuntos
Ciclo Estral/fisiologia , Regulação da Expressão Gênica/fisiologia , Genitália Feminina/metabolismo , Receptores do FSH/biossíntese , Receptores do LH/biossíntese , Receptores da Somatotropina/biossíntese , Animais , Bovinos , Feminino , Especificidade de Órgãos
3.
J Ovarian Res ; 10(1): 32, 2017 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-28472976

RESUMO

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.


Assuntos
Desidroepiandrosterona/farmacologia , Infertilidade Feminina/terapia , Reserva Ovariana/efeitos dos fármacos , Ovário/efeitos dos fármacos , Receptores Androgênicos/biossíntese , Adulto , Desidroepiandrosterona/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Fertilização in vitro/métodos , Células da Granulosa/efeitos dos fármacos , Humanos , Infertilidade Feminina/metabolismo , Infertilidade Feminina/fisiopatologia , Ovário/metabolismo , Indução da Ovulação/métodos , Gravidez , Resultado da Gravidez , Estudos Prospectivos , RNA Mensageiro/genética , Receptores Androgênicos/genética , Receptores do FSH/biossíntese , Receptores do FSH/genética , Regulação para Cima/efeitos dos fármacos
4.
Mol Reprod Dev ; 84(6): 460-467, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28337831

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Células da Granulosa/metabolismo , Interleucina-1alfa/metabolismo , Receptores do FSH/biossíntese , Animais , Feminino , Células da Granulosa/citologia , Interleucina-1alfa/genética , Camundongos , Camundongos Knockout , Receptores do FSH/genética
5.
Biomed Res Int ; 2017: 9738640, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28337462

RESUMO

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.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células da Granulosa/metabolismo , Oogênese/efeitos dos fármacos , Animais , Diferenciação Celular/genética , Família 19 do Citocromo P450/biossíntese , Estradiol/administração & dosagem , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Humanos , Técnicas de Maturação in Vitro de Oócitos , Oócitos/efeitos dos fármacos , Oócitos/crescimento & desenvolvimento , Oogênese/genética , Receptores do FSH/biossíntese , Receptores do FSH/genética , Receptores do LH/biossíntese , Receptores do LH/genética , Suínos
6.
Sci Rep ; 6: 37095, 2016 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-27848975

RESUMO

Expression of follicle-stimulation hormone receptor (FSHR) is confined to gonads and at low levels to some extragonadal tissues like human umbilical vein endothelial cells (HUVEC). FSH-FSHR signaling was shown to promote HUVEC angiogenesis and thereafter suggested to have an influential role in pregnancy. We revisited hereby the expression and functionality of FSHR in HUVECs angiogenesis, and were unable to reproduce the FSHR expression in human umbilical cord, HUVECs or immortalized HUVECs (HUV-ST). Positive controls as granulosa cells and HEK293 cells stably transfected with human FSHR cDNA expressed FSHR signal. In contrast to positive control VEGF, FSH treatment showed no effects on tube formation, nitric oxide production, wound healing or cell proliferation in HUVEC/HUV-ST. Thus, it remains open whether the FSH-FSHR activation has a direct regulatory role in the angiogenesis of HUVECs.


Assuntos
Proliferação de Células/efeitos dos fármacos , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Receptores do FSH/biossíntese , Feminino , Células HEK293 , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Gravidez , Receptores do FSH/genética
7.
Immunopharmacol Immunotoxicol ; 38(3): 197-204, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27075695

RESUMO

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.


Assuntos
Receptor alfa de Estrogênio/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análogos & derivados , Receptores do FSH/biossíntese , Receptores do LH/biossíntese , Pamoato de Triptorrelina/farmacologia , Útero/crescimento & desenvolvimento , Animais , Receptor alfa de Estrogênio/imunologia , Feminino , Regulação da Expressão Gênica/imunologia , Hormônio Liberador de Gonadotropina/farmacologia , Camundongos , Receptores do FSH/imunologia , Receptores do LH/imunologia , Útero/imunologia
8.
J Clin Endocrinol Metab ; 101(1): 254-63, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26583582

RESUMO

CONTEXT: The elevated low-density-lipoprotein cholesterol (LDL-C) in menopausal women is associated with higher risks of cardiovascular diseases. OBJECTIVE: The aim of this study is to investigate the influence and mechanism by which high postmenopausal FSH levels affect lipid profiles. METHODS: The serum FSH and lipid levels were examined in 400 Chinese postmenopausal women. The FSH receptor (FSHR) expression was identified in liver and HepG2 cells by PCR and Western blotting. The effects of FSH on lipid metabolism were confirmed in an ovariectomized mouse model by using GnRH agonist with or without additional FSH to mimic different FSH status. LDL receptor (LDLR), a necessary factor for clearance of LDL-C through endocytosis, was examined by PCR and Western blotting. RESULTS: The postmenopausal women with higher serum FSH (≥78.3 IU/L at baseline) had higher serum total cholesterol and LDL-C levels than those women with FSH levels of 40-78.3 IU/L (P < .01). The improvements of total cholesterol and LDL-C levels were more significant in higher FSH women group after treatment with hormone replacement therapy. It was only in the women whose FSH levels were reduced more than 30% after hormone replacement therapy who showed significant improvement of lipid levels. Ovariectomized mice had high serum FSH and lipids levels and reduced hepatic LDLR expression. In HepG2 cells, FSH inhibited the LDLR in a dose- and time-dependent manner, and the FSHR knockdown with specific siRNA reversed the lower LDLR induced by FSH. CONCLUSIONS: FSH may interact with its receptors in hepatocytes and reduce LDLR levels, which subsequently attenuates the endocytosis of LDL-C, resulting in an elevated circulating LDL-C level.


Assuntos
Colesterol/metabolismo , Dislipidemias/sangue , Dislipidemias/epidemiologia , Hormônio Foliculoestimulante/sangue , Fígado/metabolismo , Pós-Menopausa/sangue , Animais , Povo Asiático , Colesterol/sangue , LDL-Colesterol/sangue , Feminino , Hormônio Liberador de Gonadotropina/agonistas , Terapia de Reposição Hormonal , Humanos , Lipídeos/sangue , Fígado/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Ovariectomia , Ovário/metabolismo , Receptores do FSH/biossíntese , Receptores do FSH/genética , Receptores de LDL/biossíntese , Receptores de LDL/efeitos dos fármacos , Receptores de LDL/genética , Fatores Socioeconômicos
9.
Iran Biomed J ; 19(4): 220-5, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26175108

RESUMO

BACKGROUND: Ovarian tissue cryopreservation is an alternative strategy to preserve the fertility of women predicted to undergo premature ovarian failure. This study was designed to evaluate the expression of folliculogenesis-related genes, including factor in the germline alpha (FIGLA), growth differentiation factor-9 (GDF-9), follicle-stimulating hormone receptor (FSHR), and KIT LIGAND after vitrification/warming of human ovarian tissue. METHODS: Human ovarian tissue samples were collected from five transsexual women. In the laboratory, the ovarian medullary part was removed by a surgical blade, and the cortical tissue was cut into small pieces. Some pieces were vitrified and warmed and the others were considered as non-vitrified group (control). Follicular normality was assessed with morphological observation by a light microscope, and the expression of FIGLA, KIT LIGAND, GDF-9,, and FSHR genes was examined using real-time RT-PCR in both the vitrified and non-vitrified groups. RESULTS: Overall, 85% of the follicles preserved their normal morphologic feature after warming. The percentage of normal follicles and the expression of FIGLA, KIT LIGAND, GDF-9, and FSHR genes were similar in both vitrified and non-vitrified groups (P > 0.05). CONCLUSION: Vitrification/warming of human ovarian tissue had no remarkable effect on the expression of folliculogenesis-related genes.


Assuntos
Criopreservação/métodos , Preservação da Fertilidade/métodos , Folículo Ovariano/citologia , Folículo Ovariano/fisiologia , Vitrificação , Adulto , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Feminino , Regulação da Expressão Gênica , Fator 9 de Diferenciação de Crescimento/biossíntese , Fator 9 de Diferenciação de Crescimento/genética , Humanos , Masculino , Receptores do FSH/biossíntese , Receptores do FSH/genética , Fator de Células-Tronco/biossíntese , Fator de Células-Tronco/genética , Técnicas de Cultura de Tecidos , Pessoas Transgênero , Adulto Jovem
10.
J Biol Regul Homeost Agents ; 28(3): 419-31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25316123

RESUMO

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.


Assuntos
Aromatase/biossíntese , Proliferação de Células , Regulação da Expressão Gênica/fisiologia , Células da Granulosa/metabolismo , Receptores do FSH/biossíntese , Receptores do LH/biossíntese , Animais , Feminino , Células da Granulosa/citologia , RNA Mensageiro/biossíntese , Suínos
11.
In Vitro Cell Dev Biol Anim ; 50(8): 688-99, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24879083

RESUMO

This study aimed to evaluate the immunolocalization and messenger RNA (mRNA) expression for transforming growth factor-beta (TGF-ß) and its receptors (TGF-ßRI and RII), as well as mRNA expression for P450 aromatase and FSH receptor in caprine preantral follicles. The effects of TGF-ß, FSH alone, or in association on the in vitro follicular development were also assessed. Immunohistochemical analyses showed the expression of TGF-ß and its receptors in oocytes of all follicle stages and granulosa cells of primary and secondary follicles. mRNA for TGF-ß receptors and for FSH receptor (FSHR) was present in preantral follicles as well as in oocytes and granulosa cells of antral follicles. Isolated secondary follicles were cultured in α-minimum essential medium (MEM) alone or supplemented with either FSH (100 ng/ml), TGF-ß (10 ng/ml), or TGF-ß + FSH for 18 d. TGF-ß increased significantly oocyte diameter when compared to FSH alone and control. After 18 d of culture, all groups showed a significant reduction in P450 aromatase and FSHR mRNA levels in comparison to fresh control. In contrast, treatment with FSH significantly increased the mRNA expression for TGF-ß in comparison to fresh control and other treatments. In conclusion, the findings showed that TGF-ß and its receptors are present in caprine ovarian follicles. Furthermore, they showed a positive effect on oocyte growth in vitro.


Assuntos
Folículo Ovariano/crescimento & desenvolvimento , RNA Mensageiro/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Animais , Meios de Cultura , Feminino , Cabras , Técnicas In Vitro , Folículo Ovariano/citologia , Folículo Ovariano/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/fisiologia , Proteoglicanas/biossíntese , Proteoglicanas/fisiologia , RNA Mensageiro/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores do FSH/biossíntese , Receptores do FSH/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Fator de Crescimento Transformador beta/fisiologia
12.
J Mol Endocrinol ; 52(3): 373-82, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24711644

RESUMO

The control of mRNA translation has been mainly explored in response to activated tyrosine kinase receptors. In contrast, mechanistic details on the translational machinery are far less available in the case of ligand-bound G protein-coupled receptors (GPCRs). In this study, using the FSH receptor (FSH-R) as a model receptor, we demonstrate that part of the translational regulations occurs by phosphorylation of the translation pre-initiation complex scaffold protein, eukaryotic initiation factor 4G (eIF4G), in HEK293 cells stably expressing the FSH-R. This phosphorylation event occurred when eIF4G was bound to the mRNA 5' cap, and probably involves mammalian target of rapamycin. This regulation might contribute to cap-dependent translation in response to FSH. The cap-binding protein eIF4E also had its phosphorylation level enhanced upon FSH stimulation. We also show that FSH-induced signaling not only led to cap-dependent translation but also to internal ribosome entry site (IRES)-dependent translation of some mRNA. These data add detailed information on the molecular bases underlying the regulation of selective mRNA translation by a GPCR, and a topological model recapitulating these mechanisms is proposed.


Assuntos
Fator de Iniciação Eucariótico 4G/metabolismo , Hormônio Foliculoestimulante/metabolismo , Biossíntese de Proteínas/genética , Receptores do FSH/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Ativação Enzimática , Regulação da Expressão Gênica/genética , Células HEK293 , Humanos , Iniciação Traducional da Cadeia Peptídica/genética , Fatores de Iniciação de Peptídeos , Fosfoproteínas/metabolismo , Fosforilação , RNA Mensageiro/genética , Receptores do FSH/biossíntese , Receptores do FSH/metabolismo , Ribossomos/genética , Serina-Treonina Quinases TOR
13.
Plast Reconstr Surg ; 133(3): 344e-351e, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24572879

RESUMO

BACKGROUND: The mechanism for the growth of infantile hemangioma and vascular malformations is unknown. Follicle-stimulating hormone secretion mirrors the life cycle of infantile hemangioma and increases during adolescence, when vascular malformations often progress. The purpose of this study was to determine whether vascular anomalies express the receptor for follicle-stimulating hormone. METHODS: Human vascular tumors (i.e., infantile hemangioma, congenital hemangioma, kaposiform hemangioendothelioma, and pyogenic granuloma) and vascular malformations (i.e., capillary, lymphatic, venous, and arteriovenous) were subjected to immunofluorescence for follicle-stimulating hormone receptor. Control specimens included normal skin/subcutis, mucosa, liver, spleen, Crohn disease, granulation, pancreatitis, rheumatoid arthritis, and synovitis. Receptor and microvessel density were quantified using imaging software. RESULTS: Follicle-stimulating hormone receptor was found in the endothelium of all vascular anomalies but was not present in control specimens. Expression was greater in proliferating infantile hemangioma (6.0 percent) compared with other vascular tumors (congenital hemangioma, 0.61 percent; kaposiform hemangioendothelioma, 0.55 percent; pyogenic granuloma, 0.56 percent; p < 0.0001), despite similar microvessel density (p = 0.1). Follicle-stimulating hormone receptor was elevated in arteriovenous malformations (2.65 percent) compared with other types of vascular malformations (capillary, 1.02 percent; lymphatic, 0.38 percent; venous, 0.76 percent; p < 0.0001). CONCLUSIONS: Vascular anomalies express follicle-stimulating hormone receptor on their endothelium, in contrast to vascular control tissues. Vascular anomalies are the only benign, pathologic tissue known to express this receptor. Because the secretion of follicle-stimulating hormone correlates with the growth pattern of infantile hemangioma and vascular malformations, follicle-stimulating hormone might be involved in the pathogenesis of these lesions.


Assuntos
Receptores do FSH/biossíntese , Malformações Vasculares/metabolismo , Neoplasias Vasculares/metabolismo , Criança , Pré-Escolar , Feminino , Granuloma Piogênico/metabolismo , Hemangioma/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino
14.
Mol Hum Reprod ; 19(8): 519-27, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23562944

RESUMO

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.


Assuntos
Hormônio Antimülleriano/biossíntese , Hormônio Antimülleriano/metabolismo , Líquido Folicular/metabolismo , Células da Granulosa/metabolismo , Adolescente , Adulto , Hormônio Antimülleriano/genética , Aromatase/biossíntese , Criança , Estradiol/sangue , Feminino , Expressão Gênica , Humanos , Receptores do FSH/biossíntese , Receptores de Peptídeos/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Adulto Jovem
15.
Biomed Res Int ; 2013: 310859, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23484108

RESUMO

Antral follicular growth in the ovary is characterized by rapid expansion of granulosa cells accompanied by a rising complexity of their functionality. Within two weeks the number of human granulosa cells increases from less than 500,000 to more than 50 millions cells per follicle and differentiates into groups of cells with a variety of specialized functions involved in steroidogenesis, nursing the oocyte, and forming a functional syncitium. Both the rapid proliferation and different specialized functions of the granulosa cells can only be explained through the involvement of stem cells. However, luteinizing granulosa cells were believed to be terminally differentiated cells. Only recently, stem and progenitor cells with FSH-receptor activity were identified in populations of luteinizing granulosa cells obtained during oocyte collected for assisted reproduction. In the presence of the leukaemia-inhibiting factor (LIF), it was possible to culture a subpopulation of the luteinizing granulosa cells over prolonged time periods. Furthermore, when embedded in a matrix consisting of collagen type I, these cells continued to express the FSH receptor over prolonged time periods, developed globular formations that surrogated as follicle-like structures, providing a promising tool for reproductive biology.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Técnicas de Cultura de Células , Colágeno Tipo I/metabolismo , Matriz Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Fator Inibidor de Leucemia/metabolismo , Receptores do FSH/biossíntese
16.
Syst Biol Reprod Med ; 59(4): 191-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23394253

RESUMO

Follicle stimulating hormone receptor (FSHR) genetic variation at position 2039 A > G (rs6166, p.Asn680Ser) was repetitively shown to correspond to the measures of ovarian sensitivity and the outcomes of gonadotropin stimulation. However, to date, there has been no study revealing the mechanisms behind the associations observed. The aim of the present research was to investigate the relationship between rs6166 and mRNA expression of FSHR-dependent genes such as LHCGR, CYP19A1, and FSHR itself with particular reference to the FSHR transcript variants (deletions of exon 2, 6, and 9 and insertion of a novel exon between exons 8 and 9) in the cell culture model. Steroid production and its dependency on FSHR genotype were also assessed. A total of 22 normoovulatory patients undergoing IVF treatment were recruited. Granulosa cells were obtained by ovary puncture and cultured for 7 days to regain responsiveness to FSH in a gonadotropin-free medium. Stimulation was carried out for 24 hours in a serum-depleted environment using 0.5 UI/l rhFSH. Gene expression was assessed by real-time PCR and genotype was determined by allele-specific PCR. The distribution of p.Asn680Ser genotypes was as follows: 10 homozygotes Asn/Asn, 8 heterozygotes Asn/Ser, and 4 homozygotes Ser/Ser. Expression of total FSHR in all samples studied increased by a mean factor of 1.9 (95% CI: 0.39-11.53, p < 0.001) upon stimulation. All of the analyzed FSHR transcript variants were detectable in non-stimulated and stimulated cells. The only distinct transcript that followed up-regulation was deletion of exon 2. Homozygotes Asn/Asn tended to have higher rhFSH-induced expression of FSHR as compared to the carriers of Ser/Ser genotype. Relative expression of LHCGR and CYP19A1 although up-regulated showed no significant difference with respect to the FSHR genotype. Variable modulation of FSHR expression by its own ligand is likely to explain different clinical behavior of patients with FSHR genetic variants. The putative contribution of rs6166 requires further investigation.


Assuntos
Células da Granulosa/metabolismo , Receptores do FSH/genética , Adulto , Aromatase/biossíntese , Células Cultivadas , Feminino , Hormônio Foliculoestimulante Humano/farmacologia , Humanos , Polimorfismo Genético , Receptores do FSH/biossíntese
17.
Anim Reprod Sci ; 137(3-4): 163-76, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23375984

RESUMO

The objective of the present study was to characterize the temporal patterns of gene expression for vascular endothelial growth factors (VEGF) and VEGF receptors during ovarian follicular growth, development and maturation in buffalo (Bubalus bubalis). Follicles were classified into four groups according to size and the concentration of estradiol-17ß (E2) in follicular fluid (FF): Group I (small), 4-6mm diameter, E2>0.5ng/ml of FF; Group II (medium), 7-9mm, E2=0.5-5ng/ml; Group III (large), 10-13mm, E2=5-40ng/ml; Group IV(pre-ovulatory), >13mm, E2>180ng/ml). The mRNAs for FSH receptor (FSHR), LH receptor (LHR) and aromatase (CYP19A1) in theca interna and granulosa layers were also determined, further defining the maturational state of each group. The relative expression of VEGF isoforms (120, 164, and 188 amino acid forms), as determined by quantitative real-time PCR (qRT-PCR), increased during follicular development in both the granulosa (P<0.05) and theca layers. Relative amounts of VEGF receptors (VEGFR-1 and VEGFR-2) were least in granulosa cell (GC) and theca interna cell (TI) layers of Gp-I follicles. The amount of VEGFR-2 transcripts increased in the granulosa layer throughout development, reaching a maximum in Gp-IV follicles (P<0.05). The relative amount of VEGF isoforms and receptors in follicle lysates, as determined by western blotting, increased throughout follicular maturation to maximum amounts in pre-ovulatory follicles. Immunohistochemistry revealed a clear localization of VEGF isoforms and receptors in both steroidogenic cell types (GC and TI) and of VEGF receptors in the vascular endothelial cells of the thecal blood vessels. The most intense immunofluorescence was evident in pre-ovulatory follicles compared to other smaller follicles. These data provide evidence that the VEGF may contribute to the extensive capillary proliferation associated with the increase in size, selection, and maturation of the pre-ovulatory follicle. This may facilitate follicle maturation by enhancing the supply of nutrients, hormones, and other essential blood-borne signals to the follicle. VEGF may also promote maturation of follicles through recently recognized, non-angiogenic mechanisms.


Assuntos
Búfalos/metabolismo , Ciclo Estral/fisiologia , Folículo Ovariano/metabolismo , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Aromatase/biossíntese , Aromatase/genética , Aromatase/metabolismo , Western Blotting/veterinária , Búfalos/genética , Ciclo Estral/genética , Feminino , Regulação da Expressão Gênica , Células da Granulosa/citologia , Células da Granulosa/metabolismo , Folículo Ovariano/citologia , Isoformas de Proteínas , RNA Mensageiro/química , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Receptores do FSH/biossíntese , Receptores do FSH/genética , Receptores do FSH/metabolismo , Receptores do LH/biossíntese , Receptores do LH/genética , Receptores do LH/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/biossíntese , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Células Tecais/citologia , Células Tecais/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
18.
Gen Comp Endocrinol ; 185: 57-66, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23395683

RESUMO

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.


Assuntos
Hormônio Liberador de Gonadotropina/imunologia , Receptores do FSH/biossíntese , Receptores LHRH/biossíntese , Receptores do LH/biossíntese , Timo/metabolismo , Animais , Hormônio Foliculoestimulante/sangue , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Hormônio Luteinizante/sangue , Masculino , Ratos , Ratos Sprague-Dawley , Receptores do FSH/genética , Receptores do LH/genética , Receptores LHRH/genética , Testículo/imunologia , Testosterona/sangue , Timo/imunologia , Vacinação
19.
Res Vet Sci ; 94(3): 453-61, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23270918

RESUMO

The effects of varying concentrations of EGF were evaluated in terms of in vitro follicular development and the mRNA expression levels of EGF, EGF-R, FSH-R and P450 aromatase. After 6 days, the addition of 50 ng/mL of EGF to the culture medium increased the antrum formation rates in comparison to cultured control and after 18 days of culture produced oocytes with higher rates of meiosis resumption when compared to the other treatments (P<0.05). The daily follicular growth rates in presence of EGF (50 or 100) were increased in comparison to the cultured control (P<0.05). Treatment with EGF 50 stimulated the expression of EGF mRNA but reduced EGF-R mRNA expression and estradiol secretion as compared to the cultured control (P<0.05). After 18 days of culture, the mRNA levels for FSH-R and P450 aromatase were greater than those of the non-cultured controls (P<0.05). In conclusion, the effects of EGF treatment on the mRNA levels for EGF, EGF-R, FSH-R, and P450 aromatase varied according to the stage of follicle development.


Assuntos
Aromatase/biossíntese , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/biossíntese , Folículo Ovariano/efeitos dos fármacos , Receptores do FSH/biossíntese , Animais , Cromatina/metabolismo , Fator de Crescimento Epidérmico/biossíntese , Estradiol/análise , Estradiol/metabolismo , Feminino , Cabras , Técnicas In Vitro , Oócitos/metabolismo , Folículo Ovariano/química , Folículo Ovariano/metabolismo , Folículo Ovariano/fisiologia , Folículo Ovariano/ultraestrutura , RNA Mensageiro/metabolismo
20.
Reprod Fertil Dev ; 25(8): 1194-203, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23241220

RESUMO

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.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fator 9 de Diferenciação de Crescimento/metabolismo , Técnicas de Maturação in Vitro de Oócitos/veterinária , Oogênese , Folículo Ovariano/metabolismo , RNA Mensageiro/metabolismo , Matadouros , Animais , Bovinos , Sobrevivência Celular , Feminino , Líquido Folicular/enzimologia , Líquido Folicular/metabolismo , Glucuronosiltransferase/antagonistas & inibidores , Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Hialuronan Sintases , Isoenzimas/antagonistas & inibidores , Isoenzimas/biossíntese , Isoenzimas/genética , Isoenzimas/metabolismo , Oócitos/citologia , Oócitos/enzimologia , Oócitos/metabolismo , Folículo Ovariano/citologia , Folículo Ovariano/crescimento & desenvolvimento , Antígeno Nuclear de Célula em Proliferação/biossíntese , Antígeno Nuclear de Célula em Proliferação/química , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteoglicanas/antagonistas & inibidores , Proteoglicanas/biossíntese , Proteoglicanas/genética , Proteoglicanas/metabolismo , Receptores do FSH/antagonistas & inibidores , Receptores do FSH/biossíntese , Receptores do FSH/genética , Receptores do FSH/metabolismo , Técnicas de Cultura de Tecidos/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA