Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.206
Filter
Add more filters

Publication year range
1.
J Hum Genet ; 69(8): 365-372, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38664537

ABSTRACT

The present prospective cohort study evaluated the prevalence of FSH-R receptor Asn680Ser and Ala307Thr among infertile Indian women and the correlation of these polymorphisms with ART outcomes. Total 804 infertile and 209 fertile controls were enrolled for FSH-R analysis. Correlation of different genotypes with ovarian reserve markers, IVF parameters, and cumulative live birth rates (CLBR) was done among women undergoing IVF. In fertile controls, at 680 position GG (Ser/Ser) was the most common genotype; but among infertile women, all the genotypes were equally distributed. There was no significant difference in ovarian response parameters, oocyte yield, and CLBR among the three genotype groups. Empty follicle syndrome (EFS) was highest in women with AA or AG type at both positions. On categorisation of unexpected poor responders according to POSEIDON stratification; GG genotype at both positions had the lowest risk ratio of low-oocyte yield in ART cycles, but these differences were not statistically significant. This is the largest study from Indian ethnicity showing GG (Ser/Ser) genotype is most common among fertile women. The effect of FSH-R genotypes is very marginal on IVF parameters and is not reflected in CLBR. More prospective data may be required on the correlation of these genotypes with genuine EFS, thus stratifying the next cycles with self or donor oocytes. Routine genetic testing of FSH-R polymorphism should not be done except in a research setting. As both 680 and 307 positions are in linkage disequilibrium, only 680 position analysis may be done in a research setting.


Subject(s)
Infertility, Female , Receptors, FSH , Adult , Female , Humans , Pregnancy , Fertilization in Vitro , Genotype , India/epidemiology , Infertility, Female/genetics , Infertility, Female/epidemiology , Polymorphism, Single Nucleotide , Prevalence , Prospective Studies , Receptors, FSH/genetics , Reproductive Techniques, Assisted
2.
BMC Cancer ; 24(1): 1060, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39192214

ABSTRACT

BACKGROUND: It is challenging to improve the effects of chemotherapy and reduce its adverse impact on the ovaries. Our previous study suggested that the combination of galaxamide could enhance the antitumor effect of cisplatin (CIS) in HeLa cell xenograft mice. However, their potential effects on ovarian tissues remain unknown. METHODS: The Hela tumor-bearing female BALB/c mice model was established and randomly divided into three groups: control group (PBS group), CIS group (0.3 mg/kg CIS group) and galaxamide group (0.3 mg/kg CIS + 3 mg/kg galaxamide-treated group). The serum sex hormones levels, ovarian morphology, functional and molecular characterisation were determined and compared with those of the control group. RESULTS: The hormonal effects indicated premature ovarian insufficiency (POI) associated with CIS-induced tumor-bearing mice. CIS induces the apoptosis in primordial and developing follicles and subsequently increases follicular atresia, eventually leading to follicle loss. After cotreatment, galaxamide significantly increased anti-Mullerian hormone (AMH) and follicle-stimulating hormone receptor (FSHR) expression and prevented the CIS-induced PI3K pathway, which triggers follicle activation, apoptosis or atresia. CONCLUSION: These findings demonstrate that galaxamide could attenuate CIS-induced follicle loss by acting on the PI3K signaling pathway by stimulating AMH and/or FSHR and thus provides promising therapeutic options for patients with cervical cancer.


Subject(s)
Cisplatin , Phosphatidylinositol 3-Kinases , Primary Ovarian Insufficiency , Signal Transduction , Animals , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/metabolism , Female , Humans , Mice , Cisplatin/adverse effects , Signal Transduction/drug effects , HeLa Cells , Phosphatidylinositol 3-Kinases/metabolism , Mice, Inbred BALB C , Apoptosis/drug effects , Xenograft Model Antitumor Assays , Anti-Mullerian Hormone/blood , Anti-Mullerian Hormone/metabolism , Antineoplastic Agents/pharmacology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Receptors, FSH/metabolism , Receptors, FSH/genetics
3.
Gen Comp Endocrinol ; 354: 114542, 2024 08 01.
Article in English | MEDLINE | ID: mdl-38685391

ABSTRACT

The follicle-stimulating hormone receptor (FSHR) and luteinizing hormone receptor (LHR) in cloudy catshark were cloned, and recombinant FSHR and LHR were expressed for characterization. Ventral lobe extract (VLE) from the pituitary contains homologous FSH and LH, and it stimulated the cAMP signaling of FSHR and LHR dose-dependently. Two transcript variants of LHR (LHR-L with exon 10 and LHR-S without) were identified, and LHR-S was the dominant form with higher basal cAMP activity without VLE stimulation. Among various developmental stages of follicles, FSHR expression was mainly associated with the pre-vitellogenic and early white follicles. When follicles were recruited into vitellogenesis, the expression of FSHR decreased while of LHR was upregulated reciprocally, suggesting that LHR may also be responsible for the control of vitellogenesis in chondrichthyans. The expression of LHR-L was upregulated among maturing follicles before ovulation, indicating LHR-L could have a specific role in receiving the LH surge signal for final maturation. Plasma LH-like activity was transiently increased prior to the progesterone (P4)-surge and testosterone-drop at the beginning of P4-phase, supporting a pituitary control of follicle-maturation via LH signaling in chondrichthyans. The expression of follicular LHR was downregulated during the P4-phase when LH-like activity was high, indicating that the LH-dependent downregulation of LHR is conserved in chondrichthyans as it is in other vertebrate lineages. (213 words).


Subject(s)
Receptors, FSH , Receptors, LH , Animals , Receptors, LH/metabolism , Receptors, LH/genetics , Female , Receptors, FSH/metabolism , Receptors, FSH/genetics , Luteinizing Hormone/metabolism , Follicle Stimulating Hormone/metabolism , Fishes/metabolism , Fishes/genetics , Ovarian Follicle/metabolism
4.
Gen Comp Endocrinol ; 358: 114614, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39326529

ABSTRACT

In Gnathostomes, reproduction is mainly controlled by the hypothalamic-pituitary-gonadal (HPG) axis, with the involvement of the pituitary gonadotropic hormones (GTH), follicle-stimulating hormone (FSH) and luteinizing hormone (LH), which activate their cognate receptors, FSHR and LHR, expressed in gonads. Each GTH consists of a common α subunit and of a specific FSHß or LHß subunit. Chondrichthyes (holocephalans and elasmobranchs) is a sister group of bony vertebrates. This position is highly favorable for the understanding of the evolution of endocrine regulations of reproduction among gnathostomes. Surprisingly, the characterization of gonadotropins and their receptors is still limited in chondrichthyes. In the present study, GTH and GTHR sequences have been identified from several chondrichthyan genomes, and their primary structures were analyzed relative to human orthologs. 3D models of GTH/GTHR interaction were built, highlighting the importance of the receptor hinge region for ligand recognition. Functional hormone-receptor interactions have been studied in HEK cells using the small-spotted catshark (Scyliorhinus canicula) recombinant proteins and showed that LHR was specifically activated by LH whereas FSHR was activated by both FSH and LH. Expression profiles of GTHs and their receptors were explored by real-time PCR, in situ hybridization and immunohistochemistry during spermatogenesis, along the male genital tract and other tissues, as well as in some female tissues for comparison. Tissue-expression analyses showed that the highest levels were observed for fshr transcripts in testis and ovary and for lhr in specific extragonadal tissues. The two receptors were expressed at all stages of spermatogenesis by both germ cells and somatic cells, including undifferentiated spermatogonia, spermatocytes, spermatids, somatic precursors and Sertoli cells; differentiated Leydig cells being absent in the testis of S. canicula. Receptors were also expressed by the lymphomyeloid epigonal tissue and the testicular tubules. These results, suggest a wide range of gonadotropin-regulated functions in Elasmobranchs, as well as functional redundancy during spermatogenesis. These extended functions are discussed in an evolutionary context in which the specificity of gonadotropin signaling must have contributed to the evolution of gonadal cells' morphology and function.


Subject(s)
Gonadotropins , Receptors, Gonadotropin , Animals , Gonadotropins/metabolism , Gonadotropins/genetics , Receptors, Gonadotropin/metabolism , Receptors, Gonadotropin/genetics , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Female , Male , Humans , Luteinizing Hormone/metabolism , Phylogeny , Follicle Stimulating Hormone/metabolism
5.
Zygote ; 32(2): 130-138, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38248872

ABSTRACT

Spermatogenesis is a developmental process driven by interactions between germ cells and Sertoli cells. This process depends on appropriate gene expression, which might be regulated by transcription factors. This study focused on Rreb1, a zinc finger transcription factor, and explored its function and molecular mechanisms in spermatogenesis in a mouse model. Our results showed that RREB1 was predominantly expressed in the Sertoli cells of the testis. The decreased expression of RREB1 following injection of siRNA caused impaired Sertoli cell development, which was characterized using a defective blood-testis barrier structure and decreased expression of Sertoli cell functional maturity markers; its essential trigger might be SMAD3 destabilization. The decreased expression of RREB1 in mature Sertoli cells influenced the cell structure and function, which resulted in abnormal spermatogenesis, manifested as oligoasthenoteratozoospermia, and we believe RREB1 plays this role by regulating the transcription of Fshr and Wt1. RREB1 has been reported to activate Fshr transcription, and we demonstrated that the knockdown of Rreb1 caused a reduction in follicle-stimulating hormone receptor (FSHR) in the testis, which could be the cause of the increased sperm malformation. Furthermore, we confirmed that RREB1 directly activates Wt1 promoter activity, and RREB1 downregulation induced the decreased expression of Wt1 and its downstream polarity-associated genes Par6b and E-cadherin, which caused increased germ-cell death and reduced sperm number and motility. In conclusion, RREB1 is a key transcription factor essential for Sertoli cell development and function and is required for normal spermatogenesis.


Subject(s)
Sertoli Cells , Spermatogenesis , Transcription Factors , Animals , Male , Mice , Blood-Testis Barrier/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mice, Inbred C57BL , Receptors, FSH/genetics , Receptors, FSH/metabolism , Sertoli Cells/metabolism , Smad3 Protein/metabolism , Smad3 Protein/genetics , Spermatogenesis/genetics , Testis/metabolism , Testis/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism
6.
Pestic Biochem Physiol ; 204: 106065, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277380

ABSTRACT

Organophosphate pesticides have potent endocrine disrupting effects, hence banned in many countries. However, many organophosphates like chlorpyrifos, malathion et cetera continue to be used in some countries (Wolejko et al., 2022; Wolejko et al., 2022)including India. Fodder mediated ingestion of these substances may be harmful for livestock fertility. We have investigated the effect of the widely used organophosphate pesticide chlorpyrifos (CPF) and its metabolite, 3,5,6-trichloropyridinol (TCPy) on the expression of genes essential for spermatogenesis in goat testicular tissue. The testicular Sertoli cells (Sc) regulate germ cell division and differentiation under the influence of follicle stimulating hormone (FSH) and testosterone (T). Impaired FSH and T mediated signalling in Sc can compromise spermatogenesis leading to sub-fertility/infertility. As Sc express receptors (R) for FSH and T, they are highly susceptible to the endocrine disrupting effects of pesticides affecting fertility by dysregulating the functioning of Sc. Our results indicated that exposure to different concentrations of CPF and TCPy can compromise Sc function by downregulating the expression of FSHR and AR which was associated with a concomitant decline in the expression of genes essential for germ cell division and differentiation, like KITLG, INHBB, CLDN11 and GJA1. CPF also induced a significant reduction in the activity of acetylcholinesterase in the testes and increased the total testicular antioxidant capacity. Our results suggested that CPF and its metabolite TCPy may induce reproductive toxicity by dysregulating the expression of Sc specific genes essential for spermatogenesis.


Subject(s)
Chlorpyrifos , Goats , Spermatogenesis , Testis , Animals , Male , Spermatogenesis/drug effects , Chlorpyrifos/toxicity , Testis/drug effects , Testis/metabolism , Down-Regulation/drug effects , Insecticides/toxicity , Pyridines/pharmacology , Pyridines/toxicity , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Pyridones
7.
Women Health ; 64(4): 308-316, 2024 04.
Article in English | MEDLINE | ID: mdl-38468162

ABSTRACT

Around 70 percent of cases of Primary Ovarian Insufficiency (POI) etiology remain unexplained. The aim of our study is to contribute to the etiology and genetic background of POI. A total of 37 POI patients and 30 women in the reproductive period were included in this prospective, case-control study between August 2020 and December 2021. The women were examined for 36 genes with next-generation sequencing (NGS) panel. Gene variations were detected in 59.5 percent of the patients in the case group. FSHR p.S680N (rs6166, c.2039 G>A) and FSHR p.A307T (rs6165, c.919 G>A) gene variants, which are most frequently located in exon 10 of the FSHR gene, were detected in both groups. Although it was not found that these gene variants were significantly different between the groups, it was also found that they were significantly different in POI patients under 30 years of age and in those with a family history of POI. Variations were detected in 12 genes in POI patients. Two gene variants (FGFR1 [c.386A>C, rs765615419] and KISS1 [c.58 G>A, rs12998]) were detected in both groups, and the remaining gene variants were detected only in POI patients. No differences were detected between the groups in terms of gene variations. However, the gene variations detected only in POI patients may play a role in the etiology of POI.


Subject(s)
Genetic Variation , Primary Ovarian Insufficiency , Humans , Female , Primary Ovarian Insufficiency/genetics , Case-Control Studies , Prospective Studies , Adult , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Receptors, FSH/genetics
8.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-39000181

ABSTRACT

Perimenopause significantly impacts women's health globally, often managed with hormone replacement therapy (HRT) despite the associated risks. This study explores a novel alternative exosome therapy, aimed at stimulating estrogen production in ovarian tissues, thus offering a potential non-hormonal treatment for perimenopausal symptoms. Employing ex vivo methodologies, ovarian cortex specimens from perimenopausal women were treated with exosomes derived from human umbilical cord mesenchymal stem cells and cultured under specific conditions (patent number: PCT/US2022/073467). The exosomes were produced under cyclic guanosine monophosphate (cGMP) conditions, ensuring high safety standards. Estrogen levels were quantified using enzyme-linked immunosorbent assay (ELISA), and gene expression changes in estrogen and follicle-stimulating hormone (FSH) receptors were assessed via quantitative polymerase chain reaction (PCR). Immunohistochemistry (IHC) was utilized to evaluate cellular proliferation and apoptotic markers. The results indicated a significant increase in estrogen levels and estrogen receptor-alpha (Erα) expression in treated tissues compared to controls. Additionally, a decrease in apoptotic markers and an increase in cellular proliferation markers were observed. These findings suggest that exosome therapy can effectively enhance estrogen production and modulate receptor sensitivity in perimenopausal ovarian tissues. This approach could serve as a safer alternative to HRT, aligning with the body's natural regulatory mechanisms and potentially offering a more effective treatment option for managing perimenopausal symptoms.


Subject(s)
Estrogens , Exosomes , Perimenopause , Humans , Exosomes/metabolism , Female , Perimenopause/metabolism , Estrogens/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cell Proliferation , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/genetics , Middle Aged , Apoptosis , Receptors, FSH/metabolism , Receptors, FSH/genetics , Ovary/metabolism
9.
Zhongguo Zhong Yao Za Zhi ; 49(12): 3288-3294, 2024 Jun.
Article in Zh | MEDLINE | ID: mdl-39041091

ABSTRACT

This study aimed to explore the regulating effect of Gegen Decoction(GGD) on the hypothalamic-pituitary-ovarian axis(HPOA) dysfunction in the mouse model of primary dysmenorrhea(PD). The mouse model of PD with periodic characteristics was established by administration of estradiol benzoate and oxytocin. Mice were randomized into control, model, GGD, and ibuprofen groups. The writhing response, hypothalamus index, pituitary index, ovary index, and uterus index were observed and determined. The serum levels of prostaglandin F_(2α)(PGF_(2α)), gonadotropin-releasing hormone(GnRH), follicle-stimulating hormone(FSH), luteinizing hormone(LH), and estrogen(E_2) levels were measured by ELISA kits. Western blot and qPCR were employed to determine the protein and mRNA levels, respectively, of gonadotropin-releasing hormone receptor(GnRH-R) in the pituitary tissue, follicle-stimulating hormone receptor(FSHR) and luteinizing hormone receptor(LHR) in the ovarian tissue, and estrogen receptor(ER) in the uterine tissue. The results showed that the writhing response, serum levels of PGF_(2α), GnRH, FSH, LH, and E_2, ovarian and uterine indexes, the protein and mRNA levels of GnRH-R in the pituitary tissue, FSHR and LHR in the ovarian tissue, and ER in the uterine tissue were significantly increased in the model group compared with those in the control group. GGD inhibited the writhing response, reduced the serum levels of PGF_(2α), GnRH, FSH, LH, and E_2, decreased the ovarian and uterine indexes, and down-regulated the protein and mRNA levels of GnRH-R in the pituitary tissue, FSHR and LHR in the ovarian tissue, and ER in the uterine tissue. The data suggested that GGD can regulate the HPOA and inhibit E_2 generation in the mice experiencing recurrent PD by down-regulating the expression of proteins and genes related to HPOA axis, thus exerting the therapeutic effect on PD.


Subject(s)
Drugs, Chinese Herbal , Dysmenorrhea , Ovary , Animals , Female , Mice , Ovary/drug effects , Ovary/metabolism , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Dysmenorrhea/drug therapy , Dysmenorrhea/metabolism , Dysmenorrhea/genetics , Dysmenorrhea/physiopathology , Luteinizing Hormone/blood , Follicle Stimulating Hormone/blood , Pituitary Gland/metabolism , Pituitary Gland/drug effects , Humans , Receptors, FSH/genetics , Receptors, FSH/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism
10.
Reproduction ; 166(4): 271-284, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37590121

ABSTRACT

In brief: Exposure to cadmium (Cd) during pregnancy can potentially harm the reproductive system of male offspring. This article shows that pregnant woman should be protected from cadmium exposure. Abstract: Exposure to cadmium (Cd) during pregnancy can potentially harm the reproductive system of male offspring, although the full extent of its heritable effects remains partially unresolved. In this study, we examined the inter-generational impacts of Cd using a distinct male-lineage generational model. Pregnant Sprague-Dawley female rats (F0) were administered control or cadmium chloride (0.5, 1 and 2 mg/day) via intra-gastric administration from gestation day 1 to 20. Subsequently, the first filial generation (F1) male rats were mated with untreated females (not exposed to Cd) to produce the second filial generation (F2). Histopathological analysis of the F1 and F2 generations revealed abnormal testicular development, while ultrastructural examination indicated damage to Sertoli cells. Cd exposure also led to alterations in serum hormone levels (gonadotropin-releasing hormone, follicle-stimulating hormone) and reduced follicle-stimulating hormone receptor (FSHR) protein expression in Sertoli cells in the F1 generation. Furthermore, Cd affected the mRNA and protein expression of FSHR pathway factors and DNA methyltransferase, albeit with distinct patterns and inconsistencies observed between the F1 and F2 generations. Overall, our findings indicate that prenatal Cd exposure, using a male-lineage transmission model, can induce inter-generational effects on male reproduction, particularly by causing toxicity in Sertoli cells. This effect appears to be primarily mediated through disruptions in the FSHR pathway and changes in DNA methyltransferase activity in the male testes.


Subject(s)
Cadmium , Drug-Related Side Effects and Adverse Reactions , Female , Male , Pregnancy , Animals , Rats , Rats, Sprague-Dawley , Cadmium/toxicity , Sertoli Cells , Receptors, FSH/genetics , Methyltransferases , DNA
11.
Reprod Biomed Online ; 46(5): 826-834, 2023 05.
Article in English | MEDLINE | ID: mdl-37130623

ABSTRACT

RESEARCH QUESTION: Is there an association between FSHR sequence variants and reproductive outcomes following IVF in predicted normoresponders? DESIGN: Multicentre prospective cohort study conducted from November 2016 to June 2019 in Vietnam, Belgium and Spain including patients aged <38 years, and undergoing IVF with a predicted normal response with fixed-dose 150 IU rFSH in an antagonist protocol. Genotyping was performed for three FSHR (c.919A>G, c.2039A>G, c.-29G>A) and one FSHB sequence variants (c.-211G>T). Clinical pregnancy rate (CPR), live birth rate (LBR) and miscarriage rate in the first embryo transfer and cumulative live birth rate (CLBR) were compared between the different genotypes. RESULTS: A total of 351 patients underwent at least one embryo transfer. Genetic model analysis that adjusted for patient age, body mass index, ethnicity, type of embryo transfer, embryo stage and number of top-quality embryos transferred revealed a higher CPR for homozygous patients for the variant allele G of c.919A>G when compared to patients with genotype AA (60.3% versus 46.3%, adjusted odds ratio [ORadj] 1.96, 95% confidence interval [CI] 1.09-3.53). Also, c.919A>G genotypes AG and GG presented a higher CPR and LBR when compared with genotype AA (59.1% versus 46.3%, ORadj 1.80, 95% CI 1.08-3.00, and 51.3% versus 39.0%, ORadj 1.69, 95% CI 1.01-2.80, respectively). Cox regression models revealed a statistically significantly lower CLBR for c.2039A>G genotype GG in the codominant model (hazard ratio [HR] 0.66, 95% CI 0.43-0.99). CONCLUSION: These results demonstrate a previously unreported association between variant c.919A>G genotype GG and higher CPR and LBR in infertile patients and reinforce a potential role for genetic background in predicting the reproductive prognosis following IVF.


Subject(s)
Embryo Transfer , Receptors, FSH , Reproduction , Female , Humans , Pregnancy , Birth Rate , Embryo Transfer/methods , Fertilization in Vitro , Genotype , Live Birth , Pregnancy Rate , Prospective Studies , Retrospective Studies , Receptors, FSH/genetics
12.
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
13.
Cell Biochem Funct ; 41(6): 633-641, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37287186

ABSTRACT

Follicle-stimulating hormone receptor (FSHR) belongs to the family of G-protein coupled receptors and acts as a cognate receptor for follicle-stimulating hormone (FSH). Among the various polymorphic changes reported in FSHR, rs6165 polymorphism leading to Ala307Thr variation in the extracellular domain of the FSHR (FSHRED ) is widely reported. Therefore we attempted to evaluate the functional implications of this variation by studying its effects on FSHRED structure as well as FSH binding. Our atomic-scale investigations reveal that the hinge region, a key hormone interaction site in the extracellular domain of Wt FSHR, exhibits significantly more flexibility compared with the variant structure. Moreover, the Wt receptor in complex with FSH was observed to form a pocket-like structure in its hinge region whereas such a structure was not detected in the variant. The study further reveals that the key residue, sTyr335, required for FSH recognition and FSHR activation, exhibits lower binding free energy in the variant structure as compared to the Wt. In conclusion, our results point out that Ala307Thr variation leads to structural and conformational anomalies in FSHRED which may alter its FSH binding and affect its activation.


Subject(s)
Polycystic Ovary Syndrome , Receptors, FSH , Female , Humans , Follicle Stimulating Hormone/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Mutation , Amino Acid Substitution
14.
Cell Mol Life Sci ; 79(3): 136, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35181820

ABSTRACT

Sertoli cells (Sc) are the sole target of follicle-stimulating hormone (FSH) in the testis and attain functional maturation post-birth to significantly augment germ cell (Gc) division and differentiation at puberty. Despite having an operational microRNA (miRNA) machinery, limited information is available on miRNA-mediated regulation of Sc maturation and male fertility. We have shown before that miR-92a-3p levels decline in pubertal rat Sc. In response to FSH treatment, the expressions of FSH Receptor, Claudin11 and Klf4 were found to be elevated in pubertal rat Sc coinciding with our finding of FSH-induced decline in miR-92a-3p levels. To investigate the association of miR-92a-3p and spermatogenesis, we generated transgenic mice where such pubertal decline of miR-92a-3p was prevented by its overexpression in pubertal Sc under proximal Rhox5 promoter, which is known to be activated specifically at puberty, in Sc. Our in vivo observations provided substantial evidence that FSH-induced decline in miR-92a-3p expression during Sc maturation acts as an essential prerequisite for the pubertal onset of spermatogenesis. Elevated expression of miR-92a-3p in post-pubertal testes results into functionally compromised Sc, leading to impairment of the blood-testis barrier formation and apoptosis of pre-meiotic Gc, ultimately culminating into infertility. Collectively, our data suggest that regulation of miR-92a-3p expression is crucial for Sc-mediated induction of active spermatogenesis at puberty and regulation of male fertility.


Subject(s)
Cell Differentiation , Fertility , Follicle Stimulating Hormone/pharmacology , Germ Cells/cytology , MicroRNAs/genetics , Sertoli Cells/cytology , Testis/cytology , Animals , Female , Germ Cells/drug effects , Germ Cells/metabolism , Hormones/pharmacology , Male , Mice , Mice, Transgenic , Rats , Rats, Wistar , Receptors, FSH/genetics , Receptors, FSH/metabolism , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Sexual Maturation , Spermatogenesis , Testis/drug effects , Testis/metabolism
15.
Int J Mol Sci ; 24(2)2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36674596

ABSTRACT

After the controlled ovarian stimulation (COS), the number of cumulus oocyte complexes collected is lower than predicted. The aim of this study is to understand if there is a possible reason for that deficient ovarian response. It was hypothesized that this is associated with the SNP (single-nucleotide polymorphism) of the FSH receptor (FSHr), specifically c.2039A > G, resulting in Asn680Ser. Two groups of patients were enrolled for this purpose: the normal (n = 36) and abnormal responses (n = 31). To predict the number of retrievable oocytes, according to the anti-Mullerian hormone (AMH) and the antral follicle count (AFC), the following formula was applied in a log scale: the number of oocytes retrieved = 2.584 − 0.015 × (age) − 0.035 × (FSH) + 0.038 × (AMH) + 0.026 × (AFC). Then, when the number of oocytes collected was less than 50% of the calculated value, it was proposed that the patients result in an abnormal response. DNA sample blood was collected from the women, and then the genetic assessment for the Asn680Ser of the FSHr was evaluated in both groups. The differences between the two categories were statistically analyzed with an independent samples t test, a Mann−Whitney U test and a Chi-squared test. In a patient with an abnormal response, a significant prevalence of the amino acid serine at position 680 of the FSHr compared to the counterpart group (p < 0.05) was detected. In conclusion, according to the results, the genetic evaluation of the FSHr could represent an accurate and predictive feature for patients undergoing assisted reproductive technology treatment.


Subject(s)
Ovarian Follicle , Receptors, FSH , Female , Animals , Ovarian Follicle/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Anti-Mullerian Hormone/metabolism , Reference Values , Oocytes
16.
Int J Mol Sci ; 24(21)2023 Nov 04.
Article in English | MEDLINE | ID: mdl-37958944

ABSTRACT

Developing modulatory antibodies against G protein-coupled receptors is challenging. In this study, we targeted the follicle-stimulating hormone receptor (FSHR), a significant regulator of reproduction, with variable domains of heavy chain-only antibodies (VHHs). We built two immune VHH libraries and submitted them to multiplexed phage display approaches. We used next-generation sequencing to identify 34 clusters of specifically enriched sequences that were functionally assessed in a primary screen based on a cAMP response element (CRE)-dependent reporter gene assay. In this assay, 23 VHHs displayed negative or positive modulation of FSH-induced responses, suggesting a high success rate of the multiplexed strategy. We then focused on the largest cluster identified (i.e., PRC1) that displayed positive modulation of FSH action. We demonstrated that PRC1 specifically binds to the human FSHR and human FSHR/FSH complex while potentiating FSH-induced cAMP production and Gs recruitment. We conclude that the improved selection strategy reported here is effective for rapidly identifying functionally active VHHs and could be adapted to target other challenging membrane receptors. This study also led to the identification of PRC1, the first potential positive modulator VHH reported for the human FSHR.


Subject(s)
Bacteriophages , Receptors, FSH , Humans , Receptors, FSH/genetics , Receptors, FSH/metabolism , Follicle Stimulating Hormone/metabolism , Signal Transduction , High-Throughput Nucleotide Sequencing , Bacteriophages/genetics
17.
Mol Hum Reprod ; 28(5)2022 04 29.
Article in English | MEDLINE | ID: mdl-35471239

ABSTRACT

Follicle-stimulating hormone (FSH) is crucial in the development and regulation of reproductive functions. The actions of human FSH and its receptor (FSHR) and mutations therein have mainly been studied using in vivo models, primary cells, cancer cells and cell lines ectopically expressing the FSHR. To allow studies of endogenous FSHR function in vitro, we differentiated FSHR-expressing cells from human pluripotent stem cells. FSH stimulation of the wild-type (WT), but not the inactivating Finnish founder mutant (A189V) receptor, activated the canonical cyclic adenosine monophosphate (cAMP)-dependent signaling pathway and downstream mediators. To investigate protein-protein interaction partners of FSHR at resting state and upon FSH stimulation, we expressed FSHR in HEK293 cells followed by affinity purification mass spectrometry analyses. We found 19 specific high-confidence interacting proteins for WT FSHR and 14 for A189V FSHR, several of which have been linked to infertility. Interestingly, while only WT FSHR interacted with FSH, insulin-like growth factor 1 receptor (IGF1R), for example, interacted with both WT and A189V FSHR upon FSH stimulation. In conclusion, our protocol allows detailed studies of FSH action and disease modeling in human cells endogenously expressing FSHR.


Subject(s)
Pluripotent Stem Cells , Receptors, FSH , Follicle Stimulating Hormone/metabolism , HEK293 Cells , Humans , Mutation , Pluripotent Stem Cells/metabolism , Receptors, FSH/genetics
18.
Reprod Biomed Online ; 45(5): 943-946, 2022 11.
Article in English | MEDLINE | ID: mdl-36075849

ABSTRACT

RESEARCH QUESTION: Does the FSH receptor (FSHR) genotype influence the results of donor ovarian stimulation using corifollitropin alfa? DESIGN: A prospective cohort study was performed including 152 oocyte donor ovarian stimulations: group 1 (n = 80) using a single dose of 150 µg of corifollitropin alpha; and group 2 (n = 72) using in addition to corifollitropin alpha, continued stimulation using recombinant FSH 225 IU daily. Allelic discrimination was used to genotype the FSHR p.N680S polymorphism. Linear regression analysis was performed to study the differences between groups. RESULTS: No differences in clinical characteristics between genotypes were reported. Overall, the results of ovarian stimulation were better in oocyte donors with SN and NN genotypes compared with SS in terms of the number of retrieved oocytes (15.78 versus 10.83; P = 0.008) and retrieved metaphase II (MII) oocytes (12.34 versus 9.00; P = 0.032). Corresponding differences were also observed in group 1 for the number of retrieved oocytes (13.83 versus 7.50, P = 0.018) and retrieved MII oocytes (10.24 versus 5.42; P = 0.038). However, in group 2 no significant differences were found for oocytes retrieved (17.55 versus 13.06, P = 0.064) or MII oocytes (14.25 versus 11.39; P = 0.12). CONCLUSIONS: This study suggests that ovarian stimulation protocols with corifollitropin alfa in women with the SS genotypes could be associated with fewer oocytes and MII oocytes retrieved. Despite the fact that corifollitropin alfa has a longer half-life, the results for the SS genotype do not match those for the other genotypes, so other factors must be involved. Therefore, to tailor treatments, it would be advisable to genotype women at p.N680S of the FSHR.


Subject(s)
Follicle Stimulating Hormone , Receptors, FSH , Pregnancy , Female , Humans , Receptors, FSH/genetics , Prospective Studies , Pregnancy Rate , Follicle Stimulating Hormone, Human , Ovulation Induction/methods , Genotype
19.
Mol Biol Rep ; 49(4): 2565-2577, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35037195

ABSTRACT

BACKGROUND: The aim of this study was to study the relationship between the methylation level of the promoter region of follicle-stimulating hormone receptor (FSHR) gene and the mRNA expression of Duolang sheep fed different energy diets. METHODS: In this study, polyembryo estrus Duolang sheep under different energy levels were selected as the experimental subjects. Dietary nutrient level reference (NY/T 816-2004), medium energy level was 10.88 MJ/day, high and low energy groups were increased and decreased by 15% on the basis of medium energy level, respectively 12.51 MJ/day, 9.25 MJ/day. Through RNA and DNA extraction, qPCR, bisulfitegenomicse-quencing PCR (BSP), sequence matching and other analysis of ovarian tissue of Duolang sheep. The difference of DNA methylation level and mRNA expression of FSHR gene during estrus in Duolang sheep fed with different energy diets was detected. RESULTS: The results showed the expression level of FSHR in high energy group was significantly higher than that in low energy group (P < 0.01), the expression level of FSHR in high energy group was significantly higher than that in medium energy group (P < 0.05), and the expression level of FSHR in medium energy group was significantly higher than that in low energy group (P < 0.05). In the target fragment of the promoter region of the FSHR gene, the methylation rate was 25% in the high energy group, 50% in the normal group, and 75% in the low energy group. CONCLUSIONS: This study revealed that different dietary energy levels had certain effects on the FSHR gene DNA methylation level and mRNA expression, and the expression level was negatively correlated with methylation level.


Subject(s)
DNA Methylation , Receptors, FSH , Animals , DNA Methylation/genetics , Diet , Estrus , Female , Follicle Stimulating Hormone/metabolism , Humans , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Sheep/genetics
20.
Zygote ; 30(6): 809-818, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36134751

ABSTRACT

The follicle-stimulating hormone (FSH) and its receptor regulate the quantity and quality of spermatozoa production. Several studies have analyzed the effect of single nucleotide polymorphisms (SNPs) in exon 10 of the FSH receptor (FSHR) on basic semen parameters without yet reaching a firm consensus. The aim of this study was to evaluate the effect of p.Thr307Ala and p.Asn680Ser polymorphisms in exon 10 of the FSHR gene, in infertile men, on intracytoplasmic sperm injection (ICSI) outcomes. This study was conducted between March 2019 and February 2020 on infertile couples undergoing ICSI at Al Hadi Laboratory and Medical Center, Lebanon. Couples with severe infertility factors that may impair gametogenesis/embryogenesis (e.g. advanced maternal age, premature ovarian failure, underwent gonadotoxic treatments, etc.) were excluded from the study. Semen and blood samples were collected from infertile men on the day of oocyte collection. Infertile men (n = 173) were screened for FSHR variants using polymerase chain reaction-restriction fragment length polymorphism. Moreover, fertilization rates, embryo quality, and pregnancy outcomes were evaluated. Higher sperm concentrations were found in the p.Thr307Ala group than the p.Thr307Thr (P < 0.01) and p.Ala307Ala (P < 0.05) groups. Furthermore, fertilization rate was significantly lower in the p.Ala307Ala genotype than in the p.Thr307Thr genotype (P < 0.05). We showed that FSHR variants in infertile men undergoing ICSI could affect sperm concentration, motility, and fertilization rates. Therefore, it will be important to confirm these results in further studies using a larger sample size.


Subject(s)
Infertility , Sperm Injections, Intracytoplasmic , Pregnancy , Female , Male , Humans , Receptors, FSH/genetics , Semen , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL