Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Anticancer Agents Med Chem ; 19(14): 1719-1727, 2019.
Article in English | MEDLINE | ID: mdl-31368878

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinogenesis/drug effects , Estrogen Receptor beta/antagonists & inhibitors , Receptors, FSH/antagonists & inhibitors , Uterine Cervical Neoplasms/drug therapy , Animals , Binding Sites/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Estrogen Receptor beta/blood , Estrogen Receptor beta/metabolism , Female , Mice , Mice, Inbred Strains , Receptors, FSH/blood , Receptors, FSH/metabolism , Structure-Activity Relationship , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
2.
Biomed Res Int ; 2019: 6539294, 2019.
Article in English | MEDLINE | ID: mdl-31240219

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Nerve Growth Factor/metabolism , Primary Ovarian Insufficiency/therapy , Umbilical Cord/transplantation , Animals , Anti-Mullerian Hormone/blood , Apoptosis/drug effects , Caspase 3/blood , Cyclophosphamide/adverse effects , Estrogens/blood , Female , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/blood , Humans , Mesenchymal Stem Cells , Nerve Growth Factor/blood , Ovary/pathology , Pregnancy , Primary Ovarian Insufficiency/chemically induced , Rats , Rats, Sprague-Dawley , Receptors, FSH/blood
3.
Sci Rep ; 9(1): 4036, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858478

ABSTRACT

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.


Subject(s)
Azoospermia/congenital , Heterografts/transplantation , Spermatocytes/metabolism , Spermatogenesis/physiology , Testis/transplantation , Animals , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Male , Mice , Mice, Nude , Rats , Rats, Wistar , Receptors, FSH/blood , SOX9 Transcription Factor/metabolism , Spermatocytes/pathology , Testosterone/blood , WT1 Proteins/metabolism
4.
Horm Mol Biol Clin Investig ; 38(1)2019 Mar 25.
Article in English | MEDLINE | ID: mdl-30904901

ABSTRACT

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.


Subject(s)
Infertility, Female/genetics , Polymorphism, Single Nucleotide , Receptors, FSH/genetics , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/genetics , Adult , Female , Humans , Infertility, Female/therapy , Ovarian Reserve , Receptors, FSH/blood , Receptors, Peptide/blood , Receptors, Transforming Growth Factor beta/blood , Sperm Injections, Intracytoplasmic/statistics & numerical data
5.
Stem Cell Res Ther ; 9(1): 20, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29386068

ABSTRACT

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.


Subject(s)
Estrous Cycle/physiology , Granulosa Cells/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Ovarian Follicle/growth & development , Primary Ovarian Insufficiency/therapy , Animals , Anti-Mullerian Hormone/blood , Apoptosis/physiology , Cells, Cultured , Disease Models, Animal , Estrogens/blood , Female , Gonadotropins/blood , Granulosa Cells/cytology , Humans , Luteinizing Hormone/blood , Mice , Mice, Inbred BALB C , Placenta/cytology , Pregnancy , Primary Ovarian Insufficiency/chemically induced , Receptors, FSH/blood , Zona Pellucida Glycoproteins/administration & dosage
6.
J BUON ; 22(5): 1352-1359, 2017.
Article in English | MEDLINE | ID: mdl-29135125

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/blood , Immunohistochemistry/methods , Neoplasms/diagnosis , Ovarian Neoplasms/blood , Prostatic Neoplasms/blood , Receptors, FSH/blood , Female , Humans , Male , Neoplasms/pathology , Ovarian Neoplasms/pathology , Prostatic Neoplasms/pathology
7.
Acta Histochem ; 119(7): 727-732, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28912046

ABSTRACT

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.


Subject(s)
Receptors, FSH/genetics , Receptors, LH/genetics , Sciuridae/genetics , Seasons , Testis/metabolism , Animals , Blotting, Western , Immunohistochemistry , Male , Polymerase Chain Reaction , Receptors, FSH/blood , Receptors, LH/blood , Sciuridae/blood
8.
Theriogenology ; 87: 242-249, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27693012

ABSTRACT

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.


Subject(s)
Anti-Mullerian Hormone/blood , Cattle/physiology , Gonadotropins/blood , Inhibins/blood , Orchiectomy/veterinary , Receptors, Gonadotropin/blood , Animals , Anti-Mullerian Hormone/metabolism , Cattle/blood , Cattle/surgery , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation/physiology , Gonadotropins/metabolism , Inhibins/genetics , Inhibins/metabolism , Luteinizing Hormone/blood , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Male , Orchiectomy/methods , Receptors, FSH/blood , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, Gonadotropin/genetics , Receptors, Gonadotropin/metabolism , Receptors, LH/blood , Receptors, LH/genetics , Receptors, LH/metabolism , Scrotum/surgery
9.
Hum Reprod Update ; 22(6): 709-724, 2016 11.
Article in English | MEDLINE | ID: mdl-27566840

ABSTRACT

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.


Subject(s)
Androgens/metabolism , Anti-Mullerian Hormone/metabolism , Estradiol/metabolism , Ovarian Follicle/physiology , Polycystic Ovary Syndrome/physiopathology , Receptors, FSH/blood , Anovulation/metabolism , Aromatase/metabolism , Enzyme Activation , Female , Granulosa Cells/physiology , Humans , Ovarian Follicle/growth & development , Ovarian Hyperstimulation Syndrome/etiology , Polycystic Ovary Syndrome/metabolism , Risk Factors
10.
Hum Reprod ; 31(4): 897-904, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26905078

ABSTRACT

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.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/genetics , Ovarian Follicle/pathology , Polymorphism, Genetic , Puberty, Delayed/genetics , Receptors, FSH/genetics , Adolescent , Adult , Alleles , Child , Cohort Studies , Cross-Sectional Studies , Denmark , Estradiol/blood , Female , Follicle Stimulating Hormone, Human/blood , Follicle Stimulating Hormone, beta Subunit/blood , Follicle Stimulating Hormone, beta Subunit/metabolism , Genetic Association Studies , Humans , Inhibins/blood , Longitudinal Studies , Luteinizing Hormone/blood , Polymorphism, Single Nucleotide , Puberty, Delayed/blood , Puberty, Delayed/metabolism , Puberty, Delayed/pathology , Receptors, FSH/blood , Receptors, FSH/metabolism , Young Adult
11.
Genet Mol Res ; 13(1): 1563-9, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24668630

ABSTRACT

Follicle-stimulating hormone receptor (FSHR), which mediates the functioning of FSH, plays a central role in reproduction. We investigated bovine FSHR gene polymorphisms and analyzed their relationships with pregnancy rates after embryo transfer and with hormone concentrations on the day of embryo transfer. One reported SNP of FSHR, G-278A, located in the 5'-upstream region, was analyzed and three genotypes (GG, GA and AA) were detected in 132 Luxi cattle recipients. Statistical analysis revealed that recipients with the GG genotype had significantly higher estrogen levels on the day of embryo transfer than did GA and AA genotypes. There were no significant differences in pregnancy rates among genotypes, after embryo transfer. We conclude that variation at these loci of the FSHR gene has no significant effect on pregnancy rates in Luxi cattle.


Subject(s)
Embryo Transfer , Estrogens/blood , Receptors, FSH/genetics , Reproduction/genetics , Animals , Cattle , Female , Genotype , Polymorphism, Single Nucleotide , Pregnancy , Pregnancy Rate , Receptors, FSH/blood
12.
PLoS One ; 8(9): e75478, 2013.
Article in English | MEDLINE | ID: mdl-24058690

ABSTRACT

Spontaneous ovarian hyperstimulation syndrome (sOHSS) is a rare event that may result from a FSH-producing pituitary adenoma (FSHoma), activating mutations of the FSH receptor (FSHR), and cross-reactivity of the FSHR to elevated hCG and TSH in the setting of pregnancy or hypothyroidism. The objective of this study was to investigate whether an aberrant FSHR was present in a woman with sOHSS and a non-surgically diagnosed FSHoma whose serum FSH levels and FSH bioactivity were nearly normal. Sequencing of the patient's FSHR gene revealed a heterozygous novel missense mutation c. 1536G>A resulting in an amino acid substitution M512I. We asked whether this mutant FSHR affected FSHR-mediated signaling pathways involving cAMP/protein kinase A (PKA), phosphatidylinositol-3 kinase (PI3K)/protein kinase B (AKT) and v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog kinase (SRC)/ p42/p44 extracellular signal-regulated protein kinases (ERK1/2). Thus, 293T cells expressing wild-type (FSHRwt), the mutant FSHR (FSHRmt), or both (FSHRwt/mt) were treated with FSH and subjected to measurements of intracellular cAMP, cAMP-induced CRE (cAMP response element)-mediated luciferase assays and immunoblot analyses of phosphorylated PI3K and ERK1/2. There were no differences in luciferase activities or phosphorylation levels of ERK1/2 among FSHRwt, FSHRmt cells and FSHwt/mt cells. However, FSHRmt cells showed a significant reduction in both cAMP production and PI3K phosphorylation levels with unchanged phosphorylation of ERK1/2 upon FSH stimulation in comparison to FSHwt cells. Also, FSH treatment did not provoke PI3K phosphorylation in FSHwt/mt cells. These results indicate that the novel missense M512I FSHR mutation identified herein did not participate in hyperactivation of FSHR-mediated signaling pathways but rather in hypoactivation of the FSH-mediated PI3K/AKT pathway. Thus, this study demonstrates a new functional property of this novel mutatnt FSHR, which, however, might not be involved in the pathogenesis of sOHSS in this FSHoma patient.


Subject(s)
Mutation, Missense , Neoplasm Proteins/genetics , Ovarian Hyperstimulation Syndrome/genetics , Pituitary Neoplasms/genetics , Receptors, FSH/genetics , Adult , Amino Acid Substitution , Cell Line , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/therapeutic use , Hormone Replacement Therapy/methods , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Middle Aged , Neoplasm Proteins/blood , Ovarian Hyperstimulation Syndrome/blood , Ovarian Hyperstimulation Syndrome/drug therapy , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Pituitary Neoplasms/blood , Pituitary Neoplasms/drug therapy , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptors, FSH/blood
13.
Anim Sci J ; 84(6): 466-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23607296

ABSTRACT

We investigated the effects of gonadotropin releasing hormone (GnRH) agonist on expressions of GnRH receptor (GnRHR), follicle-stimulating hormone receptor (FSHR) and luteinizing hormone receptor (LHR) proteins in the ovaries and follicular development in the ewes. Forty-two pre-pubertal ewes were assigned to experimental groups 1 to 5 (EG-I to EG-V) and control group (CG). Ewes in EG-I, EG-II and EG-III were subcutaneously injected with 200, 300 or 400 µg alarelin antigens twice (on days 0 and 14), respectively. Ewes in EG-IV and EG-V were subcutaneously injected with 200 µg and 300 µg alarelin antigen four times (on days 0, 7, 14 and 21). Ewes in CG were subcutaneously injected with a solvent twice (on days 0 and 14). Serum concentrations of GnRH antibody in the EGs increased and were higher than (P<0.05) that of CG from day 14 to day 60. GnRH antibody concentrations in EG-IV and EG-V were higher than that in EG-I, EG-II and EG-III from days 35 to 45. Expressions of GnRHR protein in EG-IV and EG-V were lower than that in CG (P<0. 01). Expressions of FSHR and LHR proteins in EGs increased. Levels of FSHR and LHR proteins in EG-IV and EG-V (P<0.05) were higher than CG. Ovarian weights in EGs increased. Values of follicle vertical diameter, follicle transverse diameter, follicle wall thickness, follicle externatheca thickness and follicle internatheca thickness in EG-III and EG-V were greater than other groups. Primordial follicles and primary follicles developed quickly in alarelin-immunized animals. Secondary follicles and mature follicles became more abundant. Mitochondria, mitochondrial cristaes and cortical granules increased. Serum FSH concentrations of EGs remained higher than that in CG from days 28 to 70 (P<0.05). Alarelin immunization stimulated GnRH antibody production, suppressed expression of GnRHR protein, enhanced expressions of FSHR and LHR proteins in ovaries, promoted FSH secretion and thereby accelerated the development of ovaries and follicles in ewes.


Subject(s)
Gonadotropin-Releasing Hormone/agonists , Ovarian Follicle/growth & development , Ovary/chemistry , Receptors, FSH/analysis , Receptors, LHRH/analysis , Receptors, LH/analysis , Sheep/physiology , Vaccination , Animals , Antibodies/analysis , Antibody Formation , Blotting, Western , Estradiol/blood , Female , Gonadotropin-Releasing Hormone/immunology , Injections, Subcutaneous , Organ Size , Ovary/anatomy & histology , Ovary/cytology , Receptors, FSH/blood
14.
Reprod Sci ; 20(3): 211-33, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23184658

ABSTRACT

The pituitary gonadotropin follicle-stimulating hormone (FSH) interacts with its membrane-bound receptor to produce biologic effects. Traditional functions of FSH include follicular development and estradiol production in females, and the regulation of Sertoli cell action and spermatogenesis in males. Knockout mice for both the ligand (Fshb) and the receptor (Fshr) serve as models for FSH deficiency, while Fshb and Fshr transgenic mice manifest FSH excess. In addition, inactivating mutations of both human orthologs (FSHB and FSHR) have been characterized in a small number of patients, with phenotypic effects of the ligand disruption being more profound than those of its receptor. Activating human FSHR mutants have also been described in both sexes, leading to a phenotype of normal testis function (male) or spontaneous ovarian hyperstimulation syndrome (females). As determined from human and mouse models, FSH is essential for normal puberty and fertility in females, particularly for ovarian follicular development beyond the antral stage. In males, FSH is necessary for normal spermatogenesis, but there are differences in human and mouse models. The FSHB mutations in humans result in azoospermia; while FSHR mutations in humans and knockouts of both the ligand and the receptor in mice affect testicular function but do not result in absolute infertility. Available evidence also indicates that FSH may also be necessary for normal androgen synthesis in males and females.


Subject(s)
Disease Models, Animal , Follicle Stimulating Hormone/genetics , Gonadal Disorders/genetics , Mutation/genetics , Animals , Female , Follicle Stimulating Hormone/blood , Gonadal Disorders/blood , Gonadal Disorders/diagnosis , Humans , Male , Mice , Receptors, FSH/blood , Receptors, FSH/genetics
15.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 32(1): 54-7, 2012 Jan.
Article in Chinese | MEDLINE | ID: mdl-22500393

ABSTRACT

OBJECTIVE: To observe the effects of Yangjing Zhongyu Decoction (YJZYD) on the serum estradiol (E2), testosterone (T), 17-hydroxyprogesterone (17-OHP), ovarian follicle stimulating hormone receptor (FSHR), insulin-like growth factor I (IGF-1), steroid hormone acute regulator protein (StAR) mRNA expressions in female rats. METHODS: Fifty PCOS rats were equally divided into 5 groups, i. e., the control group (C, normal PNA rats), the model group (M), the low dose YJZYD group (Y1), the medium dose YJZYD group (Y2), and the high dose YJZYD group (Y3), 10 in each. The levels of serum hormones were detected using radioimmunoassay. The morphological changes of the ovary were observed using HE method. The expressions of FSHR, IGF-1, and StAR mRNA were detected using RT PCR. RESULTS: Compared with Group C, serum T and 17-OHP significantly increased (P < 0.01), E2 significantly decreased (P < 0.01), the expressions of FSHR, IGF-1, and StAR mRNA significantly decreased in Group M (P < 0.01). Compared with Group M, the serum T level significantly decreased (P < 0.01), 17-OHP decreased (P < 0.05), and E2 significantly increased in Group Y3 (P < 0.01). The expressions of FSHR, IGF-1, and StAR mRNA increased in Group Y1, Y2, and Y3. The increase was most obvious in Group Y3 (P < 0.01). CONCLUSIONS: YJZYD could lower the hyperandrogenemia of PCOS rats. It also could increase the ovarian expressions of FSHR, IGF-1, and StAR mRNA, improve the ovarian functions, and promote the follicular development.


Subject(s)
Androgens/blood , Drugs, Chinese Herbal/pharmacology , Ovary/drug effects , Polycystic Ovary Syndrome/blood , 17-alpha-Hydroxyprogesterone/blood , Animals , Estradiol/blood , Female , Insulin-Like Growth Factor I/metabolism , Ovary/metabolism , Rats , Rats, Wistar , Receptors, FSH/blood , Testosterone/blood
16.
Zoolog Sci ; 29(1): 37-42, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22233494

ABSTRACT

The effects of treatment with a combination of levonorgestrel and quinestrol (EP-1; ratio of 2:1) on reproductive hormone levels and the expression of their receptors in female Mongolian gerbils were examined. We show that serum follicle-stimulating hormone (FSH) and luteinizing hormone (LH) decreased, whereas serum estradiol (E2) and progesterone (P4) increased after EP-1 treatment. EP1 down-regulated mRNA expression of the follicle-stimulating hormone receptor (FSHR) and the estrogen receptor (ER) ßin the ovary. EP-1 up-regulated the mRNA expression of the luteinizing hormone receptor (LHR) and the progesterone receptor (PR) in the ovary as well as ERα and PR in the uterus of Mongolian gerbils. The effects were time-dependent and dose-dependent. EP-1 had no obvious effects on ERα mRNA expression in the ovary. The current study demonstrates that the effect of EP-1 on the expression of ER subtypes is tissue-specific in Mongolian gerbils. EP-1 disrupted the reproductive endocrinology of the Mongolian gerbil. These findings suggest that the effects of EP-1 on reproductive hormone levels and their receptor expression in Mongolian gerbils may be the result of synergistic actions of levonorgestrel and quinestrol, with quinestrol playing the major role.


Subject(s)
Gerbillinae/physiology , Levonorgestrel/administration & dosage , Levonorgestrel/pharmacology , Quinestrol/administration & dosage , Quinestrol/pharmacology , Animals , Contraceptive Agents, Female/administration & dosage , Contraceptive Agents, Female/pharmacology , Drug Therapy, Combination , Estradiol/blood , Estradiol/genetics , Estradiol/metabolism , Estrogens/administration & dosage , Estrogens/pharmacology , Female , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation/drug effects , Ovary/drug effects , Ovary/metabolism , Progesterone/blood , Progesterone/genetics , Progesterone/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, FSH/blood , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Reproduction
17.
Clin Chim Acta ; 412(11-12): 1048-52, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21334319

ABSTRACT

BACKGROUND: Follicle stimulating hormone receptor (FSHR), which mediates the effects of FSH, is essential for normal spermatogenesis and male reproduction. This study aimed to investigate the effects of the FSHR polymorphisms on idiopathic male infertility and serum FSH levels in Han-Chinese population. METHODS: A case-control study was conducted with 364 idiopathic infertile patients (97 nonobstructive azoospermic, 79 oligozoospermic and 188 normozoospermic) and 281 fertile controls. Three polymorphisms at nucleotide position -29 and codons 307 and 680 of the FSHR gene were genotyped by Taqman allelic discrimination and RFLP. FSH levels were measured by RIA. RESULTS: The allele and genotype frequencies of these three polymorphisms were not significantly different between each group of the cases and controls. Serum FSH concentrations did not differ between subjects with different genotypes within each group. Together the three SNPs mainly formed four discrete haplotypes. The distribution of the haplotype was not different between each group of infertile men and fertile controls and did not influence serum FSH levels in each group. CONCLUSIONS: Our findings suggest that the FSHR polymorphisms at the studied sites do not associate with idiopathic male infertility and have no influence on FSH levels both in normal and infertile males in the Han-Chinese population.


Subject(s)
Asian People/ethnology , Ethnicity/genetics , Fertility/genetics , Infertility, Male/genetics , Polymorphism, Single Nucleotide , Receptors, FSH/genetics , Adult , Asian People/genetics , Case-Control Studies , Codon/genetics , Exons/genetics , Haplotypes/genetics , Humans , Infertility, Male/blood , Male , Nucleotides/genetics , Receptors, FSH/blood
18.
BJU Int ; 108(2 Pt 2): E117-25, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21105990

ABSTRACT

OBJECTIVE: To determine whether Thr(307)-Asn(680) and Ala(307)-Ser(680) polymorphisms of the follicle-stimulating hormone receptor (FSH-R) gene are associated with male infertility, semen quality, and reproductive hormones. PATIENTS AND METHODS: The FSH-R polymorphisms at codons 680 and 307 were analysed by restriction-fragment-length polymorphism (RFLP) in 172 infertile men and in an equal number of age-matched healthy fertile men. Genotyping of the FSH-R gene was performed using the polymerase chain reaction RFLP technique. All of the participants underwent semen analysis, and reproductive hormones were also measured. RESULTS: Allelic frequencies were 29.7% serine (Ser) and 70.3% asparagine (Asn) for fertile men (the control group), and 33.1% Ser and 66.9% Asn for infertile men (P > 0.05). The FSH-R genotype at position 680 was 49.4% (Asn/Asn), 41.9% (Asn/Ser), and 8.7% (Ser/Ser) in the control group and 40.1% (Asn/Asn), 46.5% (Asn/Ser), and 13.4% (Ser/Ser) in infertile men, respectively (P > 0.05, chi-squared test). Allelic frequencies were 33.1% alanine (Ala) and 66.9% threonine (Thr) for the control group, and 37.8% Ala and 62.2% Thr for the infertile men. The frequencies of genotypes at position 307 were 45.5% Thr/Thr, 43% Thr/Ala, and 11.6% Ala/Ala for the control group and 36.1% Thr/Thr, 52.3% Thr/Ala, and 11.6% Ala/Ala for infertile men. No significant association between codon 680 and codon 307 genotypes and infertility was observed (P = 0.076 and P = 0.073, respectively). The odds ratio (OR) values indicated that individuals with the Thr/Thr + Asn/Ser combined genotypes had a > 50% decreased risk for developing infertility (OR = 0.44; 95% confidence interval [CI]: 0.22-0.77; P = 0.006). The patients with heterozygous Thr/Ala + Asn/Ser combined genotype were 2.65 times more susceptible to infertility than the control group (OR = 2.65; 95% CI: 1.74-3.82; P = 0.0053). The FSH-R codon 680 and codon 307 genotypes did not result in different serum FSH levels either in men with normal spermatogenesis (the control group) or in men with oligoasthenoteratozoospermia (infertile men). We did not observe any significant association of FSH-R genotype frequencies with any of the sperm characteristics analysed in either group. CONCLUSIONS: No significant correlation between serum FSH levels and semen characteristics, or fertility status and FSH-R gene polymorphisms was found. The combination of heterozygous Thr/Ala + Asn/Ser genotypes increases the risk for male infertility.


Subject(s)
Genetic Predisposition to Disease , Infertility, Male/genetics , Receptors, FSH/genetics , Adult , Case-Control Studies , Follicle Stimulating Hormone/blood , Genotype , Humans , Male , Polymorphism, Restriction Fragment Length , Receptors, FSH/blood , Semen
20.
J Biomed Biotechnol ; 2010: 640318, 2010.
Article in English | MEDLINE | ID: mdl-20454649

ABSTRACT

The influence of FSH receptor (FSHR) variants on male infertility is not completely understood. The present investigation is the first screening study for SNP at nucleotide position -29 in the core promoter region and codon 680 in exon 10 of the FSHR and the effect of the serum levels of FSH on male infertility in Southeast Turkey. The SNPs in codon 680 and at position -29 of the FSHR gene were analyzed by PCR-RFLP technique in 240 men with proven fathers, and 270 infertile men (150 nonobstructive azoospermic and 120 severe oligozoospermic). The separate analysis for SNP at nucleotide position -29 did not show any difference in genotypic frequencies and serum FSH levels. The genotype distribution of SNP at position 680 was different but does not influence serum FSH levels. Together the two SNPs form four discrete haplotypes (A-Thr-Asn, G-Thr-Asn, A-Ala-Ser, and G-Ala-Ser) occurring in 10 combinations. A statistically significant difference in the allelic distribution of G-Asn/G-Ser and G-Ser/G-Ser genotype between proven fathers and infertile men but there were not any statistically significant difference in the overall frequency of the four FSHR haplotypes. We conclude that the FSHR haplotype does not associate with different serum FSH levels but it is differently distributed in proven fathers and infertile men.


Subject(s)
Fathers , Infertility, Male/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, FSH/genetics , Alleles , Codon/genetics , Gene Frequency/genetics , Genotype , Humans , Male , Receptors, FSH/blood , Turkey
SELECTION OF CITATIONS
SEARCH DETAIL
...