Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Anticancer Agents Med Chem ; 19(14): 1719-1727, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31368878

RESUMO

BACKGROUND: FSH Receptor Binding Inhibitor (FRBI) blocked the binding of FSH to FSHR. Our initial study revealed FRBI reduced the maturation rate, enhanced the apoptosis of sheep Cumulus-Oocyte Complex (COCs). Little is known about whether FRBI modulates ERß and FSHR levels in the normal uterine and cancerous tissues. The present study aimed to evaluate the FRBI effects on the expressions of Estrogen Receptor-beta (ERß) and FSH receptor (FSHR) in the uteri. METHODS: 150 mice were assigned to FRBI+FSH (COM), FSH and control groups (CG). Mice of COM-1, COM-2 and COM-3 groups were simultaneously intramuscularly injected with 500, 750 and 1000 µg FRBI with 10 IU FSH, respectively for five days. Western blotting and qPCR were utilized to determine the expression of ERß and FSHR. RESULTS: In comparison with FSH group, uterine lumen and glands of COM groups became narrow. The uterine wall and endometrial epithelium were thinned, and uterine lumen became narrow. Epithelial cells were decreased. Uterine wall thicknesses of COM-1, COM-2 and COM-3 groups were reduced by 6.49%, 14.89% and 15.69% on day 30 as compared with FSH group. Uterine perimetrium thicknesses of COM-1, COM-2 and COM-3 groups were reduced by 16.17%, 17.93% and 19.92% on day 20 in comparison with FSH group. Levels of FSHR mRNAs and proteins of COM-1, COM-2 and COM-3 groups were less than FSH group on days 20 and 30 (P<0.05). ERß protein of COM-3 group was less than FSH group. Serum estradiol (E2) and FSH concentrations of COM-2 and COM-3 were lower than that of FSH group on day 30. CONCLUSION: FRBI could decrease UWT and UPT, also block the uterine development, decline expression levels of ERß and FSHR protein. Additionally, FRBI reduced the secretion of secretion of FSH and E2. Downregulating expression of FSHR and ERß may be a potential treatment regimen for cervical cancer patients.


Assuntos
Antineoplásicos/farmacologia , Carcinogênese/efeitos dos fármacos , Receptor beta de Estrogênio/antagonistas & inibidores , Receptores do FSH/antagonistas & inibidores , Neoplasias do Colo do Útero/tratamento farmacológico , Animais , Sítios de Ligação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Receptor beta de Estrogênio/sangue , Receptor beta de Estrogênio/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos , Receptores do FSH/sangue , Receptores do FSH/metabolismo , Relação Estrutura-Atividade , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia
2.
Biomed Res Int ; 2019: 6539294, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31240219

RESUMO

Chemotherapy leads to a loss of fertility and reproductive endocrine function, thereby increasing the risk of premature ovarian failure (POF). Studies have suggested that the transplantation of mesenchymal stem cells could inhibit apoptosis in ovarian granulosa cells and improve follicular development. In the present study, the effects of human umbilical cord mesenchymal stem cell (UCMSC) transplantation on ovarian function after ovarian damage caused by chemotherapy and the mechanism underlying these effects were investigated. POF model rats were obtained by the intraperitoneal injection of cyclophosphamide, and cultured UCMSCs were transplanted by tail vein injection. Serum estrogen, follicle-stimulating hormone, gonadotropin releasing hormone, and anti-Mullerian hormone levels were detected by ELISA. Folliculogenesis was evaluated by histopathological examination. The expression levels of nerve growth factor (NGF), high affinity nerve growth factor receptor (TrkA), follicle-stimulating hormone receptor (FSHR), and caspase-3 were evaluated by western blotting and RT-qPCR. The natural reproductive capacity was assessed by pregnant rate and numbers of embryos. The results indicated that UCMSC transplantation recovered disturbed hormone secretion and folliculogenesis in POF rats. NGF and TrkA levels increased, while FSHR and caspase-3 decreased. The pregnancy rate of POF rats was improved. Therefore, UCMSCs could reduce ovarian failure due to premature senescence caused by chemotherapy, and the NGF/TrkA signaling pathway was involved in the amelioration of POF.


Assuntos
Antineoplásicos/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Fator de Crescimento Neural/metabolismo , Insuficiência Ovariana Primária/terapia , Cordão Umbilical/transplante , Animais , Hormônio Antimülleriano/sangue , Apoptose/efeitos dos fármacos , Caspase 3/sangue , Ciclofosfamida/efeitos adversos , Estrogênios/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Liberador de Gonadotropina/sangue , Humanos , Células-Tronco Mesenquimais , Fator de Crescimento Neural/sangue , Ovário/patologia , Gravidez , Insuficiência Ovariana Primária/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Receptores do FSH/sangue
3.
Sci Rep ; 9(1): 4036, 2019 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-30858478

RESUMO

Testis tissue xenografting complemented with cryopreservation is a feasible technique for fertility preservation in children with malignancy receiving gonadotoxic therapy and for endangered species with high neonatal mortality rate. However, xenografted testis of human and most endangered species are known to undergo spermatogenic arrest. In this study, we xenografted immature rat testis onto immunodeficient male mice to investigate the plausible underlying causes of spermatogenic arrest. Histological analysis of xenografted testes collected 8-wk post-grafting showed incomplete spermatogenesis with pachytene-stage spermatocytes as the most advanced germ cells. Although the levels of serum luteinizing hormone and testosterone were normal in recipient mice, those of follicle stimulating hormone (FSH) were significantly high, and specific receptors of FSH were absent in the xenografts. The xenografts demonstrated dysregulated expression of Sertoli cell-transcriptional regulators (WT1 and SOX9) and secretory proteins (SCF and GDNF). In conclusion, results from our study suggested that an altered hormonal milieu in recipients and dysregulated protein expression in xenografts could be a potential cause of spermatogenic arrest in xenografted immature rat testis. Further stereological analysis of xenografts can demonstrate precise cellular composition of xenografts to decipher interactions between germ and somatic cells to better understand spermatogenic arrest in xenografted testis.


Assuntos
Azoospermia/congênito , Xenoenxertos/transplante , Espermatócitos/metabolismo , Espermatogênese/fisiologia , Testículo/transplante , Animais , Hormônio Foliculoestimulante/sangue , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Nus , Ratos , Ratos Wistar , Receptores do FSH/sangue , Fatores de Transcrição SOX9/metabolismo , Espermatócitos/patologia , Testosterona/sangue , Proteínas WT1/metabolismo
4.
Horm Mol Biol Clin Investig ; 38(1)2019 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-30904901

RESUMO

Background Infertile women may have underlying genetic abnormalities. There is, at present, a significant number of studies on the relation between the follicle stimulating hormone receptor (FSHR) or anti-Müllerian hormone type II receptor (AMHRII) polymorphisms and response to in-vitro fertilisation (IVF) treatment. However, it is not yet clear which genotype or combination of genotypes is favourable towards a better ovarian stimulation and pregnancy outcome. Materials and methods In this study we assessed the distribution of the genotypes of FSHR Ser680Asn and of AMHRII -482A>G gene polymorphisms in a group of 126 infertile women and a control group of 100 fertile women by using real-time polymerase chain reaction (RT-PCR). Results Statistical analysis showed that the frequency of the genotypes is similar in both control and IVF/ intracytoplasmic sperm injection (ICSI) groups. Further investigation of the frequency of the nine possible combinations of these polymorphisms in the groups revealed no correlation between infertility and combination of the polymorphisms. Women with one polymorphism have on average 5.5 units higher levels of AMH compared to women carrying no polymorphism. In women with no polymorphisms, for each unit of FSH increase, the average concentration of blood AMH is expected to be 72% lower. Conclusion The distribution of the FSHR Ser680Asn and of the AMHRII -482A>G gene polymorphisms, in the Greek population is similar in fertile and infertile women. The study showed that FSH and AMH correlated levels in certain cases could be used to estimate a patient's ovarian reserve.


Assuntos
Infertilidade Feminina/genética , Polimorfismo de Nucleotídeo Único , Receptores do FSH/genética , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Adulto , Feminino , Humanos , Infertilidade Feminina/terapia , Reserva Ovariana , Receptores do FSH/sangue , Receptores de Peptídeos/sangue , Receptores de Fatores de Crescimento Transformadores beta/sangue , Injeções de Esperma Intracitoplásmicas/estatística & dados numéricos
5.
Stem Cell Res Ther ; 9(1): 20, 2018 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-29386068

RESUMO

BACKGROUND: This study was performed to determine the effects of human placenta mesenchymal stem cell (hPMSC) transplantation on granulosa cell apoptosis and anti-Müllerian hormone (AMH) and follicle-stimulating hormone receptor (FSHR) expression in autoimmune drug-induced premature ovarian failure (POF) mice. The aim of this research is to investigate the mechanisms of hPMSCs on ovarian reserve capacity. METHODS: The POF mice model was established by injection of zona pellucida 3 peptide (pZP3). hPMSC transplantation was conducted by intravenous injection into mice following pZP3 treatment. The follicle number was examined by histopathology. The serum levels of FSH, LH, E2, AMH and anti-zona pellucida antibody (AzpAb) were measured by enzyme-linked immunosorbent assay. AMH and FSHR expression in the ovary was analyzed by immunohistochemistry and western blot analysis. Granulosa cell apoptosis of the ovaries was examined by In Situ Cell Death Detection Kit. Granulosa cells were isolated and treated with SiAmh interference and hPMSC supernatant to observe the effects of AMH expression on granulosa cell apoptosis in vitro. RESULTS: The results showed that hPMSC transplantation can significantly recover the estrus cycle in the POF group. Morphological staining showed that the basal follicles and sinus follicles after hPMSC transplantation were higher in POF mice than in those without treatment, and the follicle number was significantly decreased with atresia. The serum levels of FSH, LH and AzpAb in the hPMSC transplantation group were reduced considerably, but the E2 and AMH levels were significantly increased. After hPMSC transplantation, the AMH and FSHR expression in ovarian tissue was significantly higher than in the POF group as determined by immunochemistry and western blot analysis. The FSHR expression was shown in granulosa cells only, and FSHR expression increases with AMH expressed in the ovary; granulosa cell apoptosis was decreased following hPMSC transplantation. The same results were observed from the in-vitro study. CONCLUSIONS: hPMSC transplantation can significantly improve the serum levels of high gonadotropin and low estrogen of POF mice, promote follicular development, inhibit excessive follicular atresia and granulosa cell apoptosis, and improve the ovarian reserve capacity. The mechanism may be achieved by increasing the expression of AMH and FSHR in ovaries.


Assuntos
Ciclo Estral/fisiologia , Células da Granulosa/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Folículo Ovariano/crescimento & desenvolvimento , Insuficiência Ovariana Primária/terapia , Animais , Hormônio Antimülleriano/sangue , Apoptose/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Estrogênios/sangue , Feminino , Gonadotropinas/sangue , Células da Granulosa/citologia , Humanos , Hormônio Luteinizante/sangue , Camundongos , Camundongos Endogâmicos BALB C , Placenta/citologia , Gravidez , Insuficiência Ovariana Primária/induzido quimicamente , Receptores do FSH/sangue , Glicoproteínas da Zona Pelúcida/administração & dosagem
6.
J BUON ; 22(5): 1352-1359, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29135125

RESUMO

PURPOSE: Follicle-stimulating hormone receptors (FSHR) have been reported in ovarian cancer and prostate cancer cells, but recent studies have highlighted their presence in the endothelium of blood vessels belonging to multiple neoplasias. Current research attempts to determine the role of FSHR in neoplastic proliferation and possible therapeutic or diagnostic implications. This paper aimed to analyze articles that have revealed the presence and/or role of FSHR in various neoplasms in humans. METHODS: After performing an extensive search of MEDLINE/ PubMed using MeSH terms "follicle-stimulating hormone receptors" and "cancer", 22 original articles were found relevant for the subject proposed for analysis. RESULTS: FSHR were found in all neoplasms studied, being present in both tumor cells and endothelial cells of intraand perineoplasic blood vessels. Although, the presence of these receptors seemed to be ubiquitary, conclusion and the exact role of these receptors could not be stated due to heterogeneous nature of the existing studies. CONCLUSIONS: Although extensive research studies are needed in order to elucidate the exact role of FSHRs and their utility in clinical practice, joint efforts in studying their implication in neoplastic processes can lead to the use of new diagnostic and therapeutic strategies for cancer patients.


Assuntos
Biomarcadores Tumorais/sangue , Imuno-Histoquímica/métodos , Neoplasias/diagnóstico , Neoplasias Ovarianas/sangue , Neoplasias da Próstata/sangue , Receptores do FSH/sangue , Feminino , Humanos , Masculino , Neoplasias/patologia , Neoplasias Ovarianas/patologia , Neoplasias da Próstata/patologia
7.
Acta Histochem ; 119(7): 727-732, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28912046

RESUMO

The objective of this study was to evaluate whether luteinizing hormone (LH), follicle stimulating hormone (FSH) and their receptors luteinizing hormone receptor (LHR) and follicle stimulating hormone receptor (FSHR) play roles in the seasonal spermatogenesis of the wild ground squirrels. To that end, we characterized the testicular immunolocalization of LHR and FSHR, their expression on both mRNA and protein levels, as well as serum concentrations of LH and FSH in male wild ground squirrels throughout the annual reproductive cycle. Histologically, all types of spermatogenic cells including mature spermatozoa were identified in the breeding season (April), while spermatogonia and primary spermatocytes were observed in the non-breeding season (June), and spermatogonia, primary spermatocytes and secondary spermatocytes were found in pre-hibernation (September). LHR was present in Leydig cells during the whole periods with more intense staining in the breeding season; Stronger immunostaining of FSHR was observed in Sertoli cells during the breeding season compared to the non-breeding season and pre-hibernation. Consistently, the mRNA and protein levels of LHR and FSHR were higher in testes of the breeding season, and then decreased to a relatively lower level in the non-breeding season and pre-hibernation. Meanwhile, serum LH and FSH concentrations were significantly higher in the breeding season than those in the non-breeding season and pre-hibernation. These results suggested that gonadotropins and its receptors, LHR and FSHR may be involved in the regulation of seasonal changes in testicular functions of the wild ground squirrels.


Assuntos
Receptores do FSH/genética , Receptores do LH/genética , Sciuridae/genética , Estações do Ano , Testículo/metabolismo , Animais , Western Blotting , Imuno-Histoquímica , Masculino , Reação em Cadeia da Polimerase , Receptores do FSH/sangue , Receptores do LH/sangue , Sciuridae/sangue
8.
Theriogenology ; 87: 242-249, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27693012

RESUMO

Eight-week-old calves were either castrated by partial scrotal resection (SR) without removing the testes (n = 10), Burdizzo (BZ) clamp (n = 10), orchidectomy (OR; n = 10), or were left gonad intact as controls (CO; n = 10). Concentrations of anti-Muellerian hormone (AMH), inhibin A, luteinizing hormone (LH), and follicle-stimulating hormone (FSH) in plasma were determined from 16 to 48 weeks of age. At 18 months, testes of SR, BZ, and CO bulls were obtained and the immunolocalization of LH and FSH receptors and AMH analyzed. Concentration of AMH in plasma of CO and SR bulls decreased with increasing age (P < 0.001). A similar AMH profile in CO and SR indicates that SR did not induce a true cryptorchid state. In groups OR and BZ, AMH was undetectable. Plasma inhibin concentration was higher in groups CO and SR than BZ and OR (P < 0.001). Plasma LH and FSH concentrations decreased over time (P < 0.001) and were higher in groups BZ and OR than SR and CO (P < 0.001). In the testes, immunolabeling for AMH existed in Sertoli cells of CO and SR but not BZ bulls. FSH receptors were localized in Sertoli cells, Leydig cells, spermatocytes, and the epididymis of CO and SR animals, whereas LH receptors were restricted to Leydig cells. In BZ animals, FSH and LH receptors and AMH were absent, indicating complete testicular degeneration. In conclusion, AMH is a more reliable marker for the presence of testicular tissue in bulls than inhibin. Scrotal resection did not induce a true inguinal cryptorchid state but affected testicular responsiveness to gonadotropic stimulation.


Assuntos
Hormônio Antimülleriano/sangue , Bovinos/fisiologia , Gonadotropinas/sangue , Inibinas/sangue , Orquiectomia/veterinária , Receptores da Gonadotropina/sangue , Animais , Hormônio Antimülleriano/metabolismo , Bovinos/sangue , Bovinos/cirurgia , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante/genética , Hormônio Foliculoestimulante/metabolismo , Regulação da Expressão Gênica/fisiologia , Gonadotropinas/metabolismo , Inibinas/genética , Inibinas/metabolismo , Hormônio Luteinizante/sangue , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Masculino , Orquiectomia/métodos , Receptores do FSH/sangue , Receptores do FSH/genética , Receptores do FSH/metabolismo , Receptores da Gonadotropina/genética , Receptores da Gonadotropina/metabolismo , Receptores do LH/sangue , Receptores do LH/genética , Receptores do LH/metabolismo , Escroto/cirurgia
9.
Hum Reprod Update ; 22(6): 709-724, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27566840

RESUMO

BACKGROUND: Androgens, FSH, anti-Müllerian hormone (AMH) and estradiol (E2) are essential in human ovarian folliculogenesis. However, the interactions between these four players is not fully understood. OBJECTIVES AND RATIONALE: The purpose of this review is to highlight the chronological sequence of the appearance and function of androgens, FSH, AMH and E2 and to discuss controversies in the relationship between FSH and AMH. A better understanding of this interaction could supplement our current knowledge about the pathophysiology of the polycystic ovary syndrome (PCOS). SEARCH METHODS: A literature review was performed using the following search terms: androgens, FSH, FSH receptor, anti-Mullerian hormone, AMHRII, estradiol, follicle, ovary, PCOS, aromatase, granulosa cell, oocyte. The time period searched was 1980-2015 and the databases interrogated were PubMed and Web of Science. OUTCOMES: During the pre-antral ('gonadotropin-independent') follicle growth, FSH is already active and promotes follicle growth in synergy with theca cell-derived androgens. Conversely, AMH is inhibitory by counteracting FSH. We challenge the hypothesis that AMH is regulated by androgens and propose rather an indirect effect through an androgen-dependent amplification of FSH action on granulosa cells (GCs) from small growing follicles. This hypothesis implies that FSH stimulates AMH expression. During the antral ('gonadotropin-dependent') follicle growth, E2 production results from FSH-dependent activation of aromatase. Conversely, AMH is inhibitory but the decline of its expression, amplified by E2, allows full expression of aromatase, characteristic of the large antral follicles. We propose a theoretical scheme made up of two triangles that follow each other chronologically. In PCOS, pre-antral follicle growth is excessive (triangle 1) because of intrinsic androgen excess that renders GCs hypersensitive to FSH, with consequently excessive AMH expression. Antral follicle growth and differentiation are disturbed (triangle 2) because of the abnormally persisting inhibition of FSH effects by AMH that blocks aromatase. Beside anovulation, this scenario may also serve to explain the higher receptiveness to gonadotropin therapy and the increased risk of ovarian hyperstimulation syndrome (OHSS) in patients with PCOS. WIDER IMPLICATIONS: Within GCs, the balance between FSH and AMH effects is pivotal in the shift from androgen- to oestrogen-driven follicles. Our two triangles hypothesis, based on updated data from the literature, offers a pedagogic template for the understanding of folliculogenesis in the normal and polycystic ovary. It opens new avenues for the treatment of anovulation due to PCOS.


Assuntos
Androgênios/metabolismo , Hormônio Antimülleriano/metabolismo , Estradiol/metabolismo , Folículo Ovariano/fisiologia , Síndrome do Ovário Policístico/fisiopatologia , Receptores do FSH/sangue , Anovulação/metabolismo , Aromatase/metabolismo , Ativação Enzimática , Feminino , Células da Granulosa/fisiologia , Humanos , Folículo Ovariano/crescimento & desenvolvimento , Síndrome de Hiperestimulação Ovariana/etiologia , Síndrome do Ovário Policístico/metabolismo , Fatores de Risco
10.
Hum Reprod ; 31(4): 897-904, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26905078

RESUMO

STUDY QUESTION: Do variants of the genes encoding follicle stimulating hormone (FSH) beta subunit (B) and FSH receptor (R) impact circulating reproductive hormone levels and ovarian follicle maturation in healthy peripubertal girls? SUMMARY ANSWER: FSHB and FSHR genetic variants exert, alone or their combination, distinct effects on reproductive hormone levels as well as ovarian follicle maturation in healthy peripubertal girls. WHAT IS KNOWN ALREADY: FSHB and FSHR genetic variants impact reproductive hormone levels as well as associated pathologies in women. While FSHR c. 2039A>G is known to alter gonadotrophin levels in women, FSHR c.-29G>A has not yet been shown to exert effect and there are conflicting results concerning FSHB c.-211G>T. STUDY DESIGN, SIZE, DURATION: This population-based study included 633 girls recruited as part of two cohorts, the COPENHAGEN Puberty Study (2006-2014, a cross-sectional and ongoing longitudinal study) and the Copenhagen Mother-Child Cohort (1997-2002, including transabdominal ultrasound (TAUS) of the ovaries in a subset of 91 peripubertal girls). PARTICIPANTS/MATERIALS, SETTING, METHODS: Clinical examinations, including pubertal breast stage (Tanner's classification B1-B5) were performed. Circulating levels of FSH, luteinizing hormone (LH), estradiol, anti-Mullerian hormone (AMH) and inhibin-B were assessed by immunoassays. In a subset of the girls (n = 91), ovarian volume and the number/size of antral follicles were assessed by TAUS. Genotypes were determined by competitive PCR. MAIN RESULTS AND THE ROLE OF CHANCE: FSHR c.2039A>G minor alleles were positively associated with serum FSH (ß = 0.08, P = 0.004), LH (ß = 0.06, P = 0.012) and estradiol (ß = 0.06, P = 0.017) (adjusted for Tanner stages). In a combined model, FSHR c.-29G>A and FSHR c.2039A>G alleles were positively associated with FSH levels in early-pubertal girls (B2 + B3, n = 327, r = 0.1, P = 0.02) and in young adolescents (B4 + B5, n = 149, r = 0.2, P = 0.01). Serum AMH and inhibin B levels were not significantly influenced by the single nucleotide polymorphisms (SNPs). Single SNPs were not associated with follicles counts, however, a cumulative minor allele count (FSHB c.-211 G>T and FSHR c.-29G>A) was negatively associated with the number of large follicles (≥5 mm) (n = 91, P = 0.04) (adjusted for Tanner stages). LIMITATIONS, REASONS FOR CAUTION: Since we studied girls and young adolescents during pubertal transition, our study may not be fully comparable with previous studies on FSHB and FSHR variants in adult women. The group of young adolescents (Tanner B4 + B5) reflects the endocrine situation in adult women best, however, the group is not large enough to contribute substantially to the conflicting results concerning the influence of FSHB c.-211G>T in adult women. Furthermore, we have no information about the exact day of the menstrual cycle in the subgroup of girls with menarche. WIDER IMPLICATIONS OF THE FINDINGS: The sex-specific interaction of FSHB and FSHR genetic variants and physiological as well as pathological conditions is being increasingly elucidated. The variant triplet set might serve as diagnostic and pharmacogenetic marker. For the first time, we show an additional effect of FSHR c.-29G>A on serum FSH levels in healthy girls. Moreover, morphological data suggest impaired FSH-induced maturation of ovarian follicles in minor allele carriers of FSHB c.-211G>T and FSHR c.-29G>A. This may explain previous findings of delayed pubertal onset in these girls. STUDY FUNDING/COMPETING INTERESTS: Funding was provided by the Danish Agency for Science, Technology and Innovation (09-067180), Danish Ministry of the Environment, CeHoS (MST-621-00065), Capital Region of Denmark (December 2011), Ministry of Higher Education and Science (DFF-1331-00113) and EDMaRC (Danish Ministry of Health). A.S.B. was funded from December 2015 by ReproUnion (EU Interreg Öresund-Kattegat-Skagerrak). The authors declare no conflict of interest.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/genética , Folículo Ovariano/patologia , Polimorfismo Genético , Puberdade Tardia/genética , Receptores do FSH/genética , Adolescente , Adulto , Alelos , Criança , Estudos de Coortes , Estudos Transversais , Dinamarca , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante Humano/sangue , Subunidade beta do Hormônio Folículoestimulante/sangue , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Estudos de Associação Genética , Humanos , Inibinas/sangue , Estudos Longitudinais , Hormônio Luteinizante/sangue , Polimorfismo de Nucleotídeo Único , Puberdade Tardia/sangue , Puberdade Tardia/metabolismo , Puberdade Tardia/patologia , Receptores do FSH/sangue , Receptores do FSH/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA