Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Obstet Gynecol ; 143(6): e149-e152, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38574363

ABSTRACT

BACKGROUND: Since the onset of the coronavirus disease (COVID-19) pandemic, a variety of long-COVID-19 symptoms and autoimmune complications have been recognized. CASES: We report three cases of autoimmune premature poor ovarian response in patients aged 30-37 years after mild to asymptomatic COVID-19 before vaccination, with nucleotide antibody confirmation. Two patients failed to respond to maximum-dose gonadotropins for more than 4 weeks, despite a recent history of response before having COVID-19. After a month of prednisone 30 mg, these two patients had normal follicle-stimulating hormone (FSH) levels, high oocyte yield, and blastocyst formation in successful in vitro fertilization cycles. All three patients have above-average anti-müllerian hormone levels that persisted throughout their clinical ovarian insufficiency. Two patients had elevated FSH levels, perhaps resulting from FSH receptor blockade. One patient, with a history of high response to gonadotropins 75 international units per day and below-normal FSH levels, had no ovarian response to more than a month of gonadotropins (525 international units daily), suggesting autoimmune block of the FSH glycoprotein and possible FSH receptor blockade. CONCLUSION: Auto-antibody production in response to COVID-19 before vaccination may be a rare cause of autoimmune poor ovarian response. Although vaccination is likely protective, further study will be required to evaluate the effect of vaccination and duration of autoimmune FSH or FSH receptor blockade.


Subject(s)
COVID-19 , Primary Ovarian Insufficiency , Receptors, FSH , SARS-CoV-2 , Adult , Female , Humans , Betacoronavirus , Coronavirus Infections/immunology , Coronavirus Infections/complications , Coronavirus Infections/drug therapy , COVID-19/immunology , COVID-19/complications , Follicle Stimulating Hormone/blood , Pandemics , Pneumonia, Viral/immunology , Pneumonia, Viral/complications , Pneumonia, Viral/drug therapy , Primary Ovarian Insufficiency/immunology , Primary Ovarian Insufficiency/drug therapy , Receptors, FSH/antagonists & inhibitors , SARS-CoV-2/immunology
2.
Int J Biol Sci ; 18(2): 675-692, 2022.
Article in English | MEDLINE | ID: mdl-35002517

ABSTRACT

Follicle stimulating hormone (FSH) and its receptor (FSHR) have been reported to be responsible for several physiological functions and cancers. The responsiveness of stem cells and cancer stem cells towards the FSH-FSHR system make the function of FSH and its receptors more interesting in the context of cancer biology. This review is comprised of comprehensive information on FSH-FSHR signaling in normal physiology, gonadal stem cells, cancer cells, and potential options of utilizing FSH-FSHR system as an anti-cancer therapeutic target.


Subject(s)
Follicle Stimulating Hormone/metabolism , Neoplastic Stem Cells/metabolism , Receptors, FSH/metabolism , Reproduction/physiology , Animals , Follicle Stimulating Hormone/pharmacology , Humans , Neoplasms/drug therapy , Receptors, FSH/antagonists & inhibitors , Signal Transduction
3.
Biochim Biophys Acta Biomembr ; 1864(4): 183842, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34954201

ABSTRACT

Owing to the critical role of follicle stimulating hormone receptor (FSHR) signaling in human reproduction, FSHR has been widely explored for development of fertility regulators. Using high-throughput screening approaches, several low molecular weight (LMW) compounds that can modulate FSHR activity have been identified. However, the information about the binding sites of these molecules on FSHR is not known. In the present study, we extracted the structural and functional information of 161 experimentally validated LMW FSHR modulators available in PubMed records. The potential FSHR binding sites for these modulators were identified through molecular docking experiments. The binding sites were further mapped to the agonist or antagonist activity reported for these molecules in literature. MD simulations were performed to evaluate the effect of ligand binding on conformational changes in the receptor, specifically the transmembrane domain. A peptidomimetic library was screened using these binding sites. Six peptidomimetics that interacted with the residues of transmembrane domain and extracellular loops were evaluated for binding activity using in vitro cAMP assay. Two of the six peptidomimetics exhibited positive allosteric modulatory activity and four peptidomimetics exhibited negative allosteric modulatory activity. All six peptidomimetics interacted with Asp521 of hFSHR(TMD). Several of the experimentally known LMW FSHR modulators also participated in H-bond interactions with Asp521, suggesting its important role in FSHR modulatory activity.


Subject(s)
Peptidomimetics/chemistry , Receptors, FSH/agonists , Receptors, FSH/antagonists & inhibitors , Allosteric Regulation , Binding Sites , Databases, Factual , HEK293 Cells , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Library , Peptidomimetics/metabolism , Protein Domains , Receptors, FSH/metabolism
4.
Front Endocrinol (Lausanne) ; 12: 791763, 2021.
Article in English | MEDLINE | ID: mdl-34956099

ABSTRACT

An increasing number of pollutants with endocrine disrupting potential are accumulating in the environment, increasing the exposure risk for humans. Several of them are known or suspected to interfere with endocrine signals, impairing reproductive functions. Follicle-stimulating hormone (FSH) is a glycoprotein playing an essential role in supporting antral follicle maturation and may be a target of disrupting chemicals (EDs) likely impacting female fertility. EDs may interfere with FSH-mediated signals at different levels, since they may modulate the mRNA or protein levels of both the hormone and its receptor (FSHR), perturb the functioning of partner membrane molecules, modify intracellular signal transduction pathways and gene expression. In vitro studies and animal models provided results helpful to understand ED modes of action and suggest that they could effectively play a role as molecules interfering with the female reproductive system. However, most of these data are potentially subjected to experimental limitations and need to be confirmed by long-term observations in human.


Subject(s)
Endocrine Disruptors/adverse effects , Environmental Exposure/adverse effects , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Receptors, FSH/metabolism , Signal Transduction/drug effects , Animals , Endocrine Disruptors/toxicity , Female , Humans , Receptors, FSH/antagonists & inhibitors , Signal Transduction/physiology
5.
Bioorg Med Chem Lett ; 44: 128132, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34022413

ABSTRACT

In our previous study, we had identified a 9-mer peptide (FSHß (89-97)) derived from seat belt loop of human FSHß and demonstrated its ability to function as FSHR antagonist in vivo. Structure analysis revealed that the four central residues 91STDC94 within this peptide may not be critical for receptor binding. In the present study, 91STDC94 residues were substituted with alanine to generate ΔFSHß 89-97(91STDC94/AAAA) peptide. Analogous to the parent peptide, ΔFSHß 89-97(91STDC94/AAAA) peptide inhibited binding of iodinated FSH to rat FSHR and reduced FSH-induced cAMP production. The peptide could impede granulosa cell proliferation leading to reduction in FSH-mediated ovarian weight gain in immature female rats. In these rats, peptide administration further downregulated androgen receptor and estrogen receptor-alpha expression and upregulated estrogen receptor-beta expression. The results indicate that substitution of 91STDC94 with alanine did not significantly alter FSHR antagonist activity of FSHß (89-97) peptide implying that these residues are not critical for FSH-FSHR interaction and can be replaced with non-peptidic moieties for development of more potent peptidomimetics.


Subject(s)
Drug Design , Follicle Stimulating Hormone/pharmacology , Peptides/pharmacology , Peptidomimetics , Receptors, FSH/antagonists & inhibitors , Animals , Binding Sites/drug effects , Female , Follicle Stimulating Hormone/chemistry , Humans , Models, Molecular , Ovary/drug effects , Peptides/chemistry
6.
Anim Biotechnol ; 32(1): 84-91, 2021 Feb.
Article in English | MEDLINE | ID: mdl-31456468

ABSTRACT

The present study aimed to explore FSH receptor binding inhibitor (FRBI) effects on the levels of c-Myc, K-Ras and VEGF related to ovarian cancer, to evaluate the mRNA and protein levels of FSHR in the cumulus-oocyte complex (COCs). COCs were cultured for 24 h in the in vitro maturation (IVM) media replenished with 0, 10, 20, 30 and 40 µg/mL FRBI. Contents of c-Myc, K-Ras, VEGF, cAMP and IP3 in IVM media were detected with ELISA kits, respectively. The results indicated that the levels of FSHR protein and mRNA were determined with Western blotting. C-Myc contents of four FRBI + FSH-treated groups (COM groups) were reduced after IVM of COCs. C-Myc concentrations of COM-3 group was lower than the FSH group (p < .05). K-Ras and IP3 contents of COM-4 were decreased as compared to FSH group (p < .05). Expression levels of FSHR mRNAs and proteins in COM-4 group were smaller than that of FSH group. This study revealed that FRBI treatment could decrease c-Myc and K-Ras levels in the IVM medium fluids, and depress the FSHR levels of COCs. Expression levels of FSHR mRNAs and proteins of COM-4 group were significantly decreased. FRBI exerted its action via the signal pathway of IP3 and cAMP.


Subject(s)
Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, FSH/antagonists & inhibitors , Receptors, FSH/metabolism , Animals , Female , Oocytes/chemistry , Oocytes/metabolism , Sheep
7.
Peptides ; 132: 170367, 2020 10.
Article in English | MEDLINE | ID: mdl-32645381

ABSTRACT

FSH-FSHR interaction is critical for folliculogenesis, spermatogenesis and progression of several cancers. Therefore, FSHR is an attractive target for fertility regulation and cancer therapeutics. Based on homology and structural analysis of hFSH-FSHR(ECD) complex, a minimal continuous stretch within FSHß seat-belt loop (FSHß (89-97)) was identified to be crucial for FSHR interaction. The ability of FSHß (89-97) peptide to neutralize FSHR activity was evaluated by a panel of in vitro and in vivo experiments. The synthetic peptide significantly inhibited binding of [125I]-FSH to rat Fshr as well as FSH-induced cAMP production. In immature rats, FSHß (89-97) peptide administration reduced FSH-mediated increase in ovarian weight. The peptide inhibited transition of follicles from pre-antral to antral stage and hindered the cell cycle progression of granulosa cells beyond G0/G1 phase. In adult rats, administration of the peptide inhibited estradiol synthesis and significantly perturbed folliculogenesis.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/metabolism , Granulosa Cells/drug effects , Oligopeptides/pharmacology , Ovarian Follicle/drug effects , Ovary/metabolism , Receptors, FSH/antagonists & inhibitors , Animals , Crystallography, X-Ray/methods , Estradiol/biosynthesis , Female , Granulosa Cells/metabolism , Oligopeptides/chemistry , Ovarian Follicle/metabolism , Ovary/drug effects , Protein Structural Elements , Rats , Rats, Sprague-Dawley , Receptors, FSH/metabolism , Structure-Activity Relationship
8.
J Med Chem ; 62(22): 10321-10341, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31670515

ABSTRACT

The human luteinizing hormone receptor (hLH-R) is a member of the glycoprotein hormone family of G-protein-coupled receptors (GPCRs), activated by luteinizing hormone (hLH) and essentially involved in the regulation of sex hormone production. Thus, hLH-R represents a valid target for the treatment of sex hormone-dependent cancers and diseases (polycystic ovary syndrome, uterine fibroids, endometriosis) as well as contraception. Screening of the Bayer compound library led to the discovery of tetrahydrothienopyridine derivatives as novel, small-molecule (SMOL) hLH-R inhibitors and to the development of BAY-298, the first nanomolar hLH-R antagonist reducing sex hormone levels in vivo. Further optimization of physicochemical, pharmacokinetic, and safety parameters led to the identification of BAY-899 with an improved in vitro profile and proven efficacy in vivo. BAY-298 and BAY-899 serve as valuable tool compounds to study hLH-R signaling in vitro and to interfere with the production of sex hormones in vivo.


Subject(s)
Estradiol/blood , Naphthyridines/chemistry , Receptors, LH/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Dose-Response Relationship, Drug , ERG1 Potassium Channel/metabolism , Female , Granulosa Cells/drug effects , High-Throughput Screening Assays , Humans , Male , Mice , Microsomes, Liver/drug effects , Ovulation/drug effects , Ovulation/genetics , Progesterone/blood , Rats, Wistar , Receptors, FSH/antagonists & inhibitors , Receptors, LH/metabolism , Structure-Activity Relationship , Testosterone/blood
9.
Anal Biochem ; 586: 113433, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31521670

ABSTRACT

This study investigated the association of A419T (rs121909661) and T449I (rs28928870) with infertility among Iranian women and possible treatments by agonizing the mutated receptor. 151 women were genotyped at A419T and T449I sites. Homology modeling, pharmacophore modeling, virtual screening, docking and molecular dynamics (MD) were performed. A419T and T449I indicated a significant and a weak association with infertility among Iranian women (P = 0.005 and P = 0.03, respectively). Significant differences found among three genotypes of A419T with FSH (P = 0.01) and LH (P < 0.0001). G-allele carriers of A419T had susceptibility to display higher FSH and LH serum levels. In silico results revealed the most potent agonists among 3041 similar compounds and MD supported this finding. Altogether, genotyping of A419T and T449I as potential markers might be helpful in prognosis and treatment of infertility. Also, a new series of potent FSHR agonists were identified for future drug development and treatment of infertility related to FSHR dysfunction.


Subject(s)
Infertility, Female/drug therapy , Molecular Dynamics Simulation , Mutation, Missense/drug effects , Receptors, FSH/agonists , Receptors, FSH/antagonists & inhibitors , Adult , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Infertility, Female/genetics , Iran , Ligands , Male , Molecular Structure , Pregnancy , Receptors, FSH/genetics , Young Adult
10.
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
11.
Braz J Med Biol Res ; 52(7): e8381, 2019.
Article in English | MEDLINE | ID: mdl-31241714

ABSTRACT

Experiments were conducted to determine if the follicle-stimulating hormone (FSH) receptor binding inhibitor (FRBI) impacts the expression levels of AT-rich interactive domain-containing protein 1A (ARID1A) and phosphatase and tensin homolog (PTEN) in ovaries and blood, as well as expressions of follicle-stimulating hormone cognate receptor (FSHR) gene and proteins. Mice in FRBI-10, FRBI-20, FRBI-30, and FRBI-40 groups were intramuscularly injected with 10, 20, 30, and 40 mg FRBI/kg, respectively, for five consecutive days. Western blotting and qRT-PCR were utilized to determine expression levels of ARID1A and PTEN proteins and mRNAs. Serum ARID1A and PTEN concentrations of the FRBI-40 group were higher than the control group (CG) and FSH group (P<0.05). FSHR mRNA levels of FRBI-20, FRBI-30, and FRBI-40 groups were lower than that of CG and FSH groups on day 15 (P<0.05 or P<0.01). Expression levels of FSHR proteins of FRBI-30 and FRBI-40 groups were lower than those of CG and FSH groups (P<0.05). Levels of ARID1A and PTEN proteins of the FRBI-30 group were greater than CG on days 20 and 30 (P<0.05). FRBI doses had significant positive correlations to levels of ARID1A and PTEN proteins. Additionally, ARID1A and PTEN had negative correlations to FSHR mRNAs and proteins. A high dose of FRBI could promote the expression levels of ARID1A and PTEN proteins in ovarian tissues. FRBI increased serum concentrations of ARID1A and PTEN. However, FRBI depressed expression levels of FSHR mRNAs and proteins in mouse ovaries.


Subject(s)
DNA-Binding Proteins/metabolism , Follicle Stimulating Hormone/metabolism , Nuclear Proteins/blood , Ovarian Neoplasms/metabolism , PTEN Phosphohydrolase/blood , Receptors, FSH/antagonists & inhibitors , Animals , Blotting, Western , DNA-Binding Proteins/blood , Female , Mice , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Phosphorylation , Real-Time Polymerase Chain Reaction , Transcription Factors , Transcriptional Activation/genetics , Up-Regulation
12.
J Mol Graph Model ; 89: 156-166, 2019 06.
Article in English | MEDLINE | ID: mdl-30897497

ABSTRACT

BACKGROUND: The activation of follicle stimulating hormone receptor (FSHR) by FSH and the consequent downstream signaling activities are crucial for reproductive health. The role of FSHR in tumor progression as well as osteoporosis advancement has also been well established. Currently, steroid preparations of estrogen and progesterone are being used for managing fertility, in spite of the harmful side effects, as there has not been much success in identification of effective FSHR modulators. Structure-based drug design initiatives for identification of potent and specific FSHR modulators have been impeded due to the non-availability of the complete crystal structure of hFSHR complexed with FSH. METHODS: In this study, we have modeled the 3D structure of transmembrane domain (TMD) of hFSHR and identified molecules that demonstrate good binding affinity by virtual screening of drug-like library of compounds. The 3D structural and pharmacophoric features of the binders and non-binders obtained from virtual screening were further used to develop Support Vector Machine based classifier for TMD binding. Based on the observations from docking and SVM classification, a small molecule was identified for extensive MD simulations and in vitro assays for FSHR modulatory activity. RESULTS: The molecule selected based on docking score and SVM prediction was found to inhibit FSH-induced cAMP activity by 80% at 300 µM concentration. CONCLUSION: The study proposes 1,3-diphenyl-1H-pyrazole-5-carboxylate as a promising scaffold for the design of new and potent FSHR allosteric inhibitors.


Subject(s)
Computational Biology , Drug Design , Ligands , Machine Learning , Quantitative Structure-Activity Relationship , Receptors, FSH/chemistry , Binding Sites , Cell Line , Computational Biology/methods , Computers, Molecular , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Protein Binding , Receptors, FSH/antagonists & inhibitors
13.
Braz. j. med. biol. res ; 52(7): e8381, 2019. tab, graf
Article in English | LILACS | ID: biblio-1011592

ABSTRACT

Experiments were conducted to determine if the follicle-stimulating hormone (FSH) receptor binding inhibitor (FRBI) impacts the expression levels of AT-rich interactive domain-containing protein 1A (ARID1A) and phosphatase and tensin homolog (PTEN) in ovaries and blood, as well as expressions of follicle-stimulating hormone cognate receptor (FSHR) gene and proteins. Mice in FRBI-10, FRBI-20, FRBI-30, and FRBI-40 groups were intramuscularly injected with 10, 20, 30, and 40 mg FRBI/kg, respectively, for five consecutive days. Western blotting and qRT-PCR were utilized to determine expression levels of ARID1A and PTEN proteins and mRNAs. Serum ARID1A and PTEN concentrations of the FRBI-40 group were higher than the control group (CG) and FSH group (P<0.05). FSHR mRNA levels of FRBI-20, FRBI-30, and FRBI-40 groups were lower than that of CG and FSH groups on day 15 (P<0.05 or P<0.01). Expression levels of FSHR proteins of FRBI-30 and FRBI-40 groups were lower than those of CG and FSH groups (P<0.05). Levels of ARID1A and PTEN proteins of the FRBI-30 group were greater than CG on days 20 and 30 (P<0.05). FRBI doses had significant positive correlations to levels of ARID1A and PTEN proteins. Additionally, ARID1A and PTEN had negative correlations to FSHR mRNAs and proteins. A high dose of FRBI could promote the expression levels of ARID1A and PTEN proteins in ovarian tissues. FRBI increased serum concentrations of ARID1A and PTEN. However, FRBI depressed expression levels of FSHR mRNAs and proteins in mouse ovaries.


Subject(s)
Animals , Female , Rabbits , Ovarian Neoplasms/metabolism , Receptors, FSH/antagonists & inhibitors , Nuclear Proteins/blood , DNA-Binding Proteins/metabolism , PTEN Phosphohydrolase/blood , Follicle Stimulating Hormone/metabolism , Phosphorylation , Transcription Factors , Nuclear Proteins/metabolism , Transcriptional Activation/genetics , Up-Regulation , Blotting, Western , DNA-Binding Proteins/blood , PTEN Phosphohydrolase/metabolism , Real-Time Polymerase Chain Reaction
14.
Biomed Res Int ; 2018: 5032875, 2018.
Article in English | MEDLINE | ID: mdl-30112396

ABSTRACT

Mice of FRBI-1, FRBI-2, and FRBI-3 groups were intramuscularly injected with 20, 30, and 40mg/kg, respectively, for five consecutive days. Ovarian weights of three FRBI groups were reduced in comparison with FSH group. Ovarian cortex thicknesses (OCT) of the FRBI-3 group were less than that of the FSH group (P<0.05). As compared to FSH group, there were fewer numbers of secondary follicles (SFs) and mature follicles (MF) on the ovaries of FRBI-treated mice numbers of primary follicles (PFs) and SFs also decreased. In FRBI-3 mice, we found that the primordial follicles (POF) were scarcer, the follicles developed poorly, and granulosa cells became apoptosis. SF numbers of FRBI-2 and FRBI-3 groups were less than that of the FSH group on day 20 (P<0.05). Maximum longitudinal diameter (MLD) and transverse diameter (MTD) of three FRBI groups became decreased during the experiment. MLD and MTD of the FRBI-3 group were smaller than FSH group. Levels of FSHR mRNA and protein were less than that of CG and FSH group (P<0.05). ERα protein levels of FRBI group and serum concentrations of FSH and estradiol (E2) in the FRBI-treated mice were decreased when compared to CG and FSH group. In conclusion, FSH treatment could increase the numbers of SF and MF, enhance follicle development, reduce the numbers of SF and MF, and depress the follicular development of mice. Furthermore, FRBI declined the mRNA and protein levels of ERα and FSHR in the ovaries and dropped serum concentrations of FSH and E2 of mice.


Subject(s)
Estrogen Receptor alpha/metabolism , Follicle Stimulating Hormone/physiology , Ovarian Follicle/metabolism , Receptors, FSH/metabolism , Animals , Estradiol , Female , Mice , Ovary , Receptors, FSH/antagonists & inhibitors
15.
Endokrynol Pol ; 69(2): 192-198, 2018.
Article in English | MEDLINE | ID: mdl-29952426

ABSTRACT

In normal conditions follicle stimulating hormone receptors (FSHR) are expressed in zona granulosa cells of the ovary and Sertoli cells of the testis. However, the ectopic expression of FSHR was recently discovered in intra-tumoral blood vessels endothelia and/or in tumoral cells of many extra-gonadal human tumors (particularly in endocrine tumours). The paper reviews the data concerning the appearance of ectopic FSHR in particular human neoplasms. The possible involvement of FSHR in tumor progression and the use of FSHR examination for diagnostic purposes is also reviewed. Finally, the putative role of FSHR as a new target in oncological therapy is also discussed.


Subject(s)
Neoplasms/metabolism , Receptors, FSH/genetics , Blood Vessels/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasms/blood supply , Neoplasms/diagnosis , Neoplasms/drug therapy , Receptors, FSH/antagonists & inhibitors
16.
Best Pract Res Clin Endocrinol Metab ; 32(2): 189-200, 2018 04.
Article in English | MEDLINE | ID: mdl-29678285

ABSTRACT

Infertility treatment may represent a paradigmatic example of precision medicine. Follicle-stimulating hormone (FSH) has been proposed as a valuable therapeutic option both in males and in females, even if a standardized approach is far to be established. To date, several genetic mutations as well as polymorphisms have been demonstrated to significantly affect the pathophysiology of FSH-FSH receptor (FSHR) interaction, although the underlying molecular mechanisms remain unclear. This review aims to highlight possible aspects of FSH therapy that could benefit from a pharmacogenetic approach, providing an up-to-date overview of the variability of the response to FSH treatment in both sexes. Specific sections are dedicated to the clinical use of FSH in infertility and how FSHR polymorphisms may affect the therapeutic endpoints.


Subject(s)
Infertility/genetics , Infertility/therapy , Mutation , Pharmacogenetics , Receptors, FSH/genetics , Receptors, G-Protein-Coupled/genetics , Female , Follicle Stimulating Hormone/agonists , Follicle Stimulating Hormone/analogs & derivatives , Follicle Stimulating Hormone/antagonists & inhibitors , Follicle Stimulating Hormone/therapeutic use , Humans , Male , Polymorphism, Single Nucleotide , Receptors, FSH/agonists , Receptors, FSH/antagonists & inhibitors , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors
17.
J Reprod Dev ; 64(2): 129-134, 2018 Apr 13.
Article in English | MEDLINE | ID: mdl-29249775

ABSTRACT

Although the expression of gonadotropin-releasing hormone (GnRH) in the ovaries is well established, its physiological role remains unknown. The aim of this study was to determine whether ovarian GnRH mediates the actions of human chorionic gonadotropin (hCG) in the granulosa cells of immature female rats. Follicular growth was induced by administration of pregnant mare serum gonadotropin (PMSG, 15 IU/0.15 ml) on day 25 after birth, and hCG (20 IU/0.2 ml) was administered on day 27 revealing the increase of plasma progesterone level. Primary cultures of granulosa cells were established from large follicles 2 days after PMSG treatment. Progesterone synthesis was augmented by hCG in a dose-dependent manner. Annexin A5 (ANXA5), a biomarker of GnRH, was expressed in the granulosa-luteal cells after hCG treatment, as shown by immunohistochemistry, suggesting that hCG treatment induced GnRH action. The GnRH mRNA level was increased by hCG, and treatment with GnRH agonist (GnRHa) increased ANXA5 mRNA levels in the primary cultures of granulosa cells. Concomitant incubation of GnRH (10-7 M) or GnRHa (fertirelin acetate, 10-8 M) with hCG suppressed progesterone synthesis during a 3 h incubation period. The mRNA expression of luteinizing hormone receptor (LHR) and follicle-stimulating hormone receptor (FSHR) was synergistically stimulated and suppressed by hCG and GnRHa, respectively. GnRHa stimulated p21 expression, and GnRHa and hCG synergistically reduced the mRNA expression levels of p27 and FOXO1. These data suggest that GnRH induced by LH may have a role for the LH-mediated luteinization of granulosa cells. In addition, ANXA5 may be involved in GnRH action. GnRH-ANXA5 would be an important mechanism in cell differentiation.


Subject(s)
Chorionic Gonadotropin/pharmacology , Fertility Agents, Female/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gonadotropin-Releasing Hormone/agonists , Granulosa Cells/drug effects , Luteinization/drug effects , Ovary/drug effects , Animals , Annexin A5/agonists , Annexin A5/genetics , Annexin A5/metabolism , Biomarkers/blood , Biomarkers/metabolism , Cells, Cultured , Female , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropins, Equine/pharmacology , Granulosa Cells/cytology , Granulosa Cells/metabolism , Humans , Immunohistochemistry , Ovary/cytology , Ovary/growth & development , Ovary/metabolism , Progesterone/agonists , Progesterone/antagonists & inhibitors , Progesterone/biosynthesis , Progesterone/blood , Rats, Wistar , Receptors, FSH/agonists , Receptors, FSH/antagonists & inhibitors , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, LH/agonists , Receptors, LH/genetics , Receptors, LH/metabolism
18.
Biochem Biophys Res Commun ; 493(4): 1567-1572, 2017 12 02.
Article in English | MEDLINE | ID: mdl-29017919

ABSTRACT

Antibodies are essential reagents that are increasingly used in diagnostics and therapy. Their specificity and capacity to recognize their native antigen are critical characteristics for their in vivo application. Follicle-stimulating hormone receptor is a GPCR protein regulating ovarian follicular maturation and spermatogenesis. Recently, its potentiality as a cancer biomarker has been demonstrated but no antibody suitable for in vivo tumor targeting and treatment has been characterized so far. In this paper we describe the first successful attempt to recover recombinant antibodies against the FSHR and that: i) are directly panned from a pre-immune library using whole cells expressing the target receptor at their surface; ii) show inhibitory activity towards the FSH-induced cAMP accumulation; iii) do not share the same epitope with the natural binder FSH; iv) can be produced inexpensively as mono- or bivalent functional molecules in the bacterial cytoplasm. We expect that the proposed biopanning strategy will be profitable to identify useful functional antibodies for further members of the GPCR class.


Subject(s)
Peptide Library , Receptors, FSH/antagonists & inhibitors , Receptors, FSH/immunology , Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Animals , Antibody Specificity , Cyclic AMP/metabolism , Female , Follicle Stimulating Hormone/pharmacology , HEK293 Cells , Humans , Immunization , L Cells , Male , Mice , Protein Domains , Receptors, FSH/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Signal Transduction , Solubility
19.
Nature ; 546(7656): 107-112, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28538730

ABSTRACT

Menopause is associated with bone loss and enhanced visceral adiposity. A polyclonal antibody that targets the ß-subunit of the pituitary hormone follicle-stimulating hormone (Fsh) increases bone mass in mice. Here, we report that this antibody sharply reduces adipose tissue in wild-type mice, phenocopying genetic haploinsufficiency for the Fsh receptor gene Fshr. The antibody also causes profound beiging, increases cellular mitochondrial density, activates brown adipose tissue and enhances thermogenesis. These actions result from the specific binding of the antibody to the ß-subunit of Fsh to block its action. Our studies uncover opportunities for simultaneously treating obesity and osteoporosis.


Subject(s)
Adipose Tissue/metabolism , Adiposity , Follicle Stimulating Hormone, beta Subunit/antagonists & inhibitors , Thermogenesis , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Adipose Tissue, Beige/drug effects , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Animals , Antibodies/immunology , Antibodies/pharmacology , Diet, High-Fat/adverse effects , Female , Follicle Stimulating Hormone, beta Subunit/immunology , Haploinsufficiency , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Obesity/drug therapy , Obesity/prevention & control , Osteoporosis/drug therapy , Ovariectomy , Oxygen Consumption/drug effects , Receptors, FSH/antagonists & inhibitors , Receptors, FSH/genetics , Receptors, FSH/metabolism , Thermogenesis/drug effects , Uncoupling Protein 1/biosynthesis
20.
Reproduction ; 150(2): 151-63, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25948249

ABSTRACT

The aim of the present study was to determine the direct cause of the mutation-induced, increased ovulation rate in Booroola Merino (BB) sheep. Granulosa cells were removed from antral follicles before ovulation and post-ovulation from BB (n=5) and WT (n=12) Merino ewes. Direct immunofluorescence measurement of mature cell surface receptors using flow cytometry demonstrated a significant up-regulation of FSH receptor (FSHR), transforming growth factor beta type 1, bone morphogenetic protein receptor (BMPR1B), and LH receptor (LHR) in BB sheep. The increased density of FSHR and LHR provide novel evidence of a mechanism for increasing the number of follicles that are recruited during dominant follicle selection. The compounding increase in receptors with increasing follicle size maintained the multiple follicles and reduced the apoptosis, which contributed to a high ovulation rate in BB sheep. In addition, we report a mutation-independent mechanism of down-regulation to reduce receptor density of the leading dominant follicle in sheep. The suppression of receptor density coincides with the cessation of mitogenic growth and steroidogenic differentiation as part of the luteinization of the follicle. The BB mutation-induced attenuation of BMPR1B signaling led to an increased density of the FSHR and LHR and a concurrent reduction in apoptosis to increase the ovulation rate. The role of BMPs in receptor modulation is implicated in the development of multiple ovulations.


Subject(s)
Apoptosis/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Granulosa Cells/drug effects , Ovulation/genetics , Receptors, FSH/genetics , Receptors, LH/genetics , Animals , Apoptosis/drug effects , Apoptosis/physiology , Bone Morphogenetic Protein Receptors, Type I/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type I/metabolism , Female , Flow Cytometry , Mutation/genetics , Mutation/physiology , Ovarian Follicle/anatomy & histology , Ovarian Follicle/physiology , Ovulation/drug effects , Ovulation/physiology , Pregnancy , Receptors, FSH/antagonists & inhibitors , Receptors, FSH/metabolism , Receptors, LH/antagonists & inhibitors , Receptors, LH/metabolism , Sheep, Domestic , Steroids/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL