Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.146
Filter
1.
Commun Biol ; 7(1): 844, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987655

ABSTRACT

Estrogen excess in females has been linked to a diverse array of chronic and acute diseases. Emerging research shows that exposure to estrogen-like compounds such as bisphenol S leads to increases in 17ß-estradiol levels, but the mechanism of action is unclear. The aim of this study was to reveal the underlying signaling pathway-mediated mechanisms, target site and target molecule of action of bisphenol S causing excessive estrogen synthesis. Human ovarian granulosa cells SVOG were exposed to bisphenol S at environmentally relevant concentrations (1 µg/L, 10 µg/L, and 100 µg/L) for 48 h. The results confirms that bisphenol S accumulates mainly on the cell membrane, binds to follicle stimulating hormone receptor (FSHR) located on the cell membrane, and subsequently activates the downstream cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling pathway, leading to enhanced conversion of testosterone to 17ß-estradiol. This study deepens our knowledge of the mechanisms of environmental factors in pathogenesis of hyperestrogenism.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Cyclic AMP , Estrogens , Phenols , Receptors, FSH , Signal Transduction , Sulfones , Phenols/toxicity , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Cyclic AMP/metabolism , Signal Transduction/drug effects , Female , Estrogens/metabolism , Receptors, FSH/metabolism , Receptors, FSH/genetics , Sulfones/pharmacology , Estradiol/metabolism , Granulosa Cells/metabolism , Granulosa Cells/drug effects
2.
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
3.
Zhongguo Zhong Yao Za Zhi ; 49(12): 3288-3294, 2024 Jun.
Article in Chinese | 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
4.
Cells ; 13(14)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39056753

ABSTRACT

Androgen excess is a key feature of several clinical phenotypes of polycystic ovary syndrome (PCOS). However, the presence of FSH receptor (FSHR) and aromatase (CYP19A1) activity responses to physiological endocrine stimuli play a critical role in the pathogenesis of PCOS. Preliminary data suggest that myo-Inositol (myo-Ins) and D-Chiro-Inositol (D-Chiro-Ins) may reactivate CYP19A1 activity. We investigated the steroidogenic pathway of Theca (TCs) and Granulosa cells (GCs) in an experimental model of murine PCOS induced in CD1 mice exposed for 10 weeks to a continuous light regimen. The effect of treatment with different combinations of myo-Ins and D-Chiro-Ins on the expression of Fshr, androgenic, and estrogenic enzymes was analyzed by real-time PCR in isolated TCs and GCs and in ovaries isolated from healthy and PCOS mice. Myo-Ins and D-Chiro-Ins, at a ratio of 40:1 at pharmacological and physiological concentrations, positively modulate the steroidogenic activity of TCs and the expression of Cyp19a1 and Fshr in GCs. Moreover, in vivo, inositols (40:1 ratio) significantly increase Cyp19a1 and Fshr. These changes in gene expression are mirrored by modifications in hormone levels in the serum of treated animals. Myo-Ins and D-Chiro-Ins in the 40:1 formula efficiently rescued PCOS features by up-regulating aromatase and FSHR levels while down-regulating androgen excesses produced by TCs.


Subject(s)
Aromatase , Disease Models, Animal , Inositol , Ovary , Polycystic Ovary Syndrome , Receptors, FSH , Female , Animals , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/drug therapy , Inositol/pharmacology , Mice , Aromatase/metabolism , Aromatase/genetics , Receptors, FSH/metabolism , Receptors, FSH/genetics , Ovary/metabolism , Ovary/drug effects , Ovary/pathology , Granulosa Cells/metabolism , Granulosa Cells/drug effects , Theca Cells/metabolism , Theca Cells/drug effects , Steroids/biosynthesis
5.
Theriogenology ; 226: 110-119, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38875921

ABSTRACT

The increased LH levels resulting from the absence of negative feedback after castration has been linked to long-term health issues. A need exists for an alternative contraceptive agent that functions without interfering the LH pathways. This study aimed to develop antibody fragments against the follicular-stimulating hormone receptor (anti-FSHr) using phage-display technology and evaluate its effects on Sertoli cell functions. Phage clones against the extracellular domain of dog and cat FSHr selected from an antibody fragment phagemid library were analyzed for binding kinetics by surface plasmon resonance. Sertoli cells were isolated from testes of adult animals (five dogs and five cats). Efficacy test was performed by treating Sertoli cell cultures (SCCs) with anti-FSHr antibody fragments compared with untreated in triplicates. Expressions of androgen binding protein (ABP), inhibin subunit beta B (IHBB) and vascular endothelial growth factor A (VEGFA) mRNA in SCCs were quantified by RT-qPCR. The results demonstrated that the molecular weight of the purified dog and cat anti-FSHr antibody fragment was 25 kDa and 15 kDa, respectively. Based on protein molecular weight, the antibody fragment of dogs and cats was therefore, so-called single-chain variable fragments (scFv) and nanobody (nb), respectively. The binding affinity with dissociation constant (KD) was 2.32 × 10-7 M and 2.83 × 10-9 M for dog and cat anti-FSHr antibody fragments, respectively. The cross-binding kinetic interactions between the dog anti-FSHr scFv and the cat ECD of FSHr could not be fitted to the curves to determine the binding kinetics. However, the cross-binding affinity KD between the cat anti-FSHr nb and the dog ECD FSHr was 1.75 × 10-4 M. The mRNA expression of ABP, IHBB and VEGFA in SCCs was less (P < 0.05) in both dogs (12.26, 4.07 and 5.11 folds, respectively) and cats (39.53, 14.07 and 20.29 folds, respectively) treated with anti-FSHr antibody fragments, indicating the Sertoli cell functions were suppressed. In conclusion, this study demonstrated the establishment of species-specific antibody fragments against FSHr in SCCs for dogs and cats. The fragment proteins illustrate potential to be developed as non-surgical contraceptive agent targeting FSHr in companion animals.


Subject(s)
Receptors, FSH , Animals , Dogs , Cats , Male , Receptors, FSH/metabolism , Receptors, FSH/genetics , Receptors, FSH/immunology , Contraception/veterinary , Contraception/methods , Sertoli Cells/metabolism
6.
Sci Rep ; 14(1): 10176, 2024 05 03.
Article in English | MEDLINE | ID: mdl-38702476

ABSTRACT

Experimental evidence indicates that follicle-stimulating hormone (FSH), an essential hormone for reproduction, can act directly on endothelial cells inducing atherosclerosis activation and development. However, it remains unknown whether the FSH-receptor (FSHR) is expressed in human atherosclerosis plaques. To demonstrate the FSHR presence, we used immunohistochemical and immunoelectron microscopy involving a specific monoclonal antibody FSHR1A02 that recognizes an epitope present in the FSHR-ectodomain. In all 55 patients with atherosclerotic plaques located in carotid, coronary, femoral arteries, and iliac aneurysm, FSHR was selectively expressed in arterial endothelium covering atherosclerotic plaques and endothelia lining intraplaque neovessels. Lymphatic neovessels were negative for FSHR. M1-macrophages, foam cells, and giant multinucleated cells were also FSHR-positive. FSHR was not detected in normal internal thoracic artery. Immunoelectron microscopy performed in ApoEKO/hFSHRKI mice with atherosclerotic plaques, after injection in vivo with mouse anti-hFSHR monoclonal antibody FSHR1A02 coupled to colloidal gold, showed FSHR presence on the luminal surface of arterial endothelial cells covering atherosclerotic plaques. Therefore, FSHR can bind, internalize, and deliver into the plaque circulating ligands to FSHR-positive cells. In conclusion, we report FSHR expression in endothelial cells, M1-macrophages, M1-derived foam cells, giant multinucleated macrophages, and osteoclasts associated with human atherosclerotic plaques.


Subject(s)
Plaque, Atherosclerotic , Receptors, FSH , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Humans , Receptors, FSH/metabolism , Animals , Mice , Female , Male , Macrophages/metabolism , Aged , Middle Aged , Endothelial Cells/metabolism , Foam Cells/metabolism , Foam Cells/pathology
7.
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
8.
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
9.
Drug Discov Ther ; 18(2): 106-116, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38631868

ABSTRACT

Limited studies are associated with premature ovarian insufficiency (POI)-related osteoimmune disorder currently. Bu-Shen-Ning-Xin decoction (BSNXD) displayed a favorable role in treating postmenopausal osteoporosis. However, its impact on the POI-related osteoimmune disorder remains unclear. The study primarily utilized animal experiments and network pharmacology to investigate the effects and underlying mechanisms of BSNXD on the POI-related osteoimmune disorder. First, a 4-vinylcyclohexene dioxide (VCD)-induced POI murine model was conducted to explore the therapeutical action of BSNXD. Second, we analyzed the active compounds of BSNXD and predicted their potential mechanisms for POI-related osteoimmune disorder via network pharmacology, further confirmed by molecular biology experiments. The results demonstrated that VCD exposure led to elevated follicle-stimulating hormone (FSH) levels, a 50% reduction in the primordial follicles, bone microstructure changes, and macrophage activation, indicating an osteoimmune disorder. BSNXD inhibited macrophage activation and osteoclast differentiation but did not affect serum FSH and estradiol levels in the VCD-induced POI model. Network pharmacology predicted the potential mechanisms of BSNXD against the POI-related osteoimmune disorder involving tumor necrosis factor α and MAPK signaling pathways, highlighting BSNXD regulated inflammation, hormone, and osteoclast differentiation. Further experiments identified BSNXD treatment suppressed macrophage activation via downregulating FSH receptor (FSHR) expression and inhibiting the phosphorylation of ERK and CCAAT enhancer binding proteins ß. In conclusion, BSNXD regulated POI-related osteoimmune disorder by suppressing the FSH/FSHR pathway to reduce macrophage activation and further inhibiting osteoclastogenesis.


Subject(s)
Disease Models, Animal , Drugs, Chinese Herbal , Follicle Stimulating Hormone , Macrophage Activation , Primary Ovarian Insufficiency , Receptors, FSH , Primary Ovarian Insufficiency/drug therapy , Primary Ovarian Insufficiency/chemically induced , Animals , Female , Mice , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Macrophage Activation/drug effects , Follicle Stimulating Hormone/blood , Receptors, FSH/metabolism , Vinyl Compounds/pharmacology , Vinyl Compounds/therapeutic use , Network Pharmacology , Cyclohexenes/pharmacology , Cyclohexenes/therapeutic use , Signal Transduction/drug effects , Humans , Osteoclasts/drug effects , Osteoclasts/metabolism , Cell Differentiation/drug effects
10.
Mol Cell Endocrinol ; 589: 112235, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38621656

ABSTRACT

Luteinizing hormone (LH) is essential for reproduction, controlling ovulation and steroidogenesis. Its receptor (LHR) recruits various transducers leading to the activation of a complex signaling network. We recently identified iPRC1, the first variable fragment from heavy-chain-only antibody (VHH) interacting with intracellular loop 3 (ICL3) of the follicle-stimulating hormone receptor (FSHR). Because of the high sequence similarity of the human FSHR and LHR (LHCGR), here we examined the ability of the iPRC1 intra-VHH to modulate LHCGR activity. In this study, we demonstrated that iPRC1 binds LHCGR, to a greater extent when the receptor was stimulated by the hormone. In addition, it decreased LH-induced cAMP production, cAMP-responsive element-dependent transcription, progesterone and testosterone production. These impairments are not due to Gs nor ß-arrestin recruitment to the LHCGR. Consequently, iPRC1 is the first intra-VHH to bind and modulate LHCGR biological activity, including steroidogenesis. It should help further understand signaling mechanisms elicited at this receptor and their outcomes on reproduction.


Subject(s)
Luteinizing Hormone , Receptors, LH , Signal Transduction , Receptors, LH/metabolism , Receptors, LH/genetics , Humans , Signal Transduction/drug effects , Luteinizing Hormone/metabolism , Animals , Cyclic AMP/metabolism , Protein Binding , Progesterone/metabolism , Receptors, FSH/metabolism , Receptors, FSH/genetics , Testosterone/metabolism , Testosterone/biosynthesis , HEK293 Cells , GTP-Binding Proteins/metabolism , Steroids/biosynthesis , Steroids/metabolism
11.
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
12.
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
13.
Endocrinology ; 165(4)2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38298132

ABSTRACT

Early puberty poses a significant challenge for male Atlantic salmon in aquaculture due to its negative impact on growth and welfare. The regulation of puberty in vertebrates involves 2 key reproductive hormones: follicle-stimulating hormone (FSH) and luteinizing hormone (LH) and their gonadal receptors. In male mice lacking FSH receptor, testes size is reduced, but fertility is maintained, while medaka and zebrafish with a disrupted fshr gene exhibit near normal testis size and fertility. In these fishes both Fsh and Lh are present during puberty and Lh may rescue fertility, while in salmonid fish only Fsh is present in the circulation during puberty. Using CRISPR-Cas9, we produced crispants with a high prevalence of fshr mutations at the target site, which remained fertile, although more than half showed a testis development deviating from wild-type (wt) males. Crossing out these F0 crispants to each other produced a viable F1 generation showing frameshift (fshr-/-) or in-frame mutations (fshrif/if). Nearly all wt males matured while all fshr-/- males remained immature with small testes containing A spermatogonia as the furthest developed germ cell type and prepubertal plasma androgen levels. Also, the pituitary transcript levels of gnrhr2bba and lhb, but not for fshb, were reduced in the fshr-/- males compared with maturing males. More than half of the fshrif/if mutant males showed no or a delayed maturation. In conclusion, Atlantic salmon show the unique characteristic that loss of Fshr function alone results in male infertility, offering new opportunities to control precocious puberty or fertility in salmon.


Subject(s)
Receptors, FSH , Salmo salar , Male , Animals , Mice , Receptors, FSH/genetics , Receptors, FSH/metabolism , Salmo salar/genetics , Salmo salar/metabolism , Zebrafish/genetics , Sexual Maturation/genetics , Follicle Stimulating Hormone/metabolism , Testis/metabolism
14.
PLoS Comput Biol ; 20(1): e1011415, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38206994

ABSTRACT

Glycoprotein hormone receptors [thyrotropin (TSHR), luteinizing hormone/chorionic gonadotropin (LHCGR), and follicle stimulating hormone (FSHR) receptors] are rhodopsin-like G protein-coupled receptors. These receptors display common structural features including a prominent extracellular domain with leucine-rich repeats (LRR) stabilized by ß-sheets and a long and flexible loop known as the hinge region (HR), and a transmembrane (TM) domain with seven α-helices interconnected by intra- and extracellular loops. Binding of the ligand to the LRR resembles a hand coupling transversally to the α- and ß-subunits of the hormone, with the thumb being the HR. The structure of the FSH-FSHR complex suggests an activation mechanism in which Y335 at the HR binds into a pocket between the α- and ß-chains of the hormone, leading to an adjustment of the extracellular loops. In this study, we performed molecular dynamics (MD) simulations to identify the conformational changes of the FSHR and LHCGR. We set up a FSHR structure as predicted by AlphaFold (AF-P23945); for the LHCGR structure we took the cryo-electron microscopy structure for the active state (PDB:7FII) as initial coordinates. Specifically, the flexibility of the HR domain and the correlated motions of the LRR and TM domain were analyzed. From the conformational changes of the LRR, TM domain, and HR we explored the conformational landscape by means of MD trajectories in all-atom approximation, including a membrane of polyunsaturated phospholipids. The distances and procedures here defined may be useful to propose reaction coordinates to describe diverse processes, such as the active-to-inactive transition, and to identify intermediaries suited for allosteric regulation and biased binding to cellular transducers in a selective activation strategy.


Subject(s)
Follicle Stimulating Hormone , Molecular Dynamics Simulation , Amino Acid Sequence , Cryoelectron Microscopy , Receptors, FSH/chemistry , Receptors, FSH/metabolism , Lipids
15.
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
16.
FEBS Lett ; 598(2): 220-232, 2024 01.
Article in English | MEDLINE | ID: mdl-37923554

ABSTRACT

Intracellular variable fragments of heavy-chain antibody from camelids (intra-VHH) have been successfully used as chaperones to solve the 3D structure of active G protein-coupled receptors bound to their transducers. However, their effect on signalling has been poorly explored, although they may provide a better understanding of the relationships between receptor conformation and activity. Here, we isolated and characterized iPRC1, the first intra-VHH recognizing a member of the large glycoprotein hormone receptor family, the follicle-stimulating hormone receptor (FSHR). This intra-VHH recognizes the FSHR third intracellular loop and decreases cAMP production in response to FSH, without altering Gαs recruitment. Hence, iPRC1 behaves as an allosteric modulator and provides a new tool to complete structure/activity studies performed thus far on this receptor.


Subject(s)
Follicle Stimulating Hormone , Receptors, FSH , Receptors, FSH/genetics , Receptors, FSH/chemistry , Receptors, FSH/metabolism , Follicle Stimulating Hormone/chemistry , Follicle Stimulating Hormone/metabolism , Follicle Stimulating Hormone/pharmacology , GTP-Binding Proteins/metabolism , Signal Transduction
17.
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
18.
Nat Commun ; 14(1): 6991, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37914684

ABSTRACT

Follicle-stimulating hormone (FSH) is involved in mammalian reproduction via binding to FSH receptor (FSHR). However, several studies have found that FSH and FSHR play important roles in extragonadal tissue. Here, we identified the expression of FSHR in human and mouse pancreatic islet ß-cells. Blocking FSH signaling by Fshr knock-out led to impaired glucose tolerance owing to decreased insulin secretion, while high FSH levels caused insufficient insulin secretion as well. In vitro, we found that FSH orchestrated glucose-stimulated insulin secretion (GSIS) in a bell curve manner. Mechanistically, FSH primarily activates Gαs via FSHR, promoting the cAMP/protein kinase A (PKA) and calcium pathways to stimulate GSIS, whereas high FSH levels could activate Gαi to inhibit the cAMP/PKA pathway and the amplified effect on GSIS. Our results reveal the role of FSH in regulating pancreatic islet insulin secretion and provide avenues for future clinical investigation and therapeutic strategies for postmenopausal diabetes.


Subject(s)
Follicle Stimulating Hormone , Islets of Langerhans , Mice , Animals , Humans , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/metabolism , Insulin Secretion , Glucose/pharmacology , Glucose/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Islets of Langerhans/metabolism , Signal Transduction , Insulin/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Mammals/metabolism
19.
Vitam Horm ; 123: 555-585, 2023.
Article in English | MEDLINE | ID: mdl-37717998

ABSTRACT

Follicle-stimulating hormone (FSH) and estrogens are fundamental to support reproductive functions. Beside the well-known FSH membrane receptor (FSHR), a G protein-coupled estrogen receptor (GPER) has been found, over the last two decades, in several tissues. It may trigger rapid, non-genomic responses of estradiol, activating proliferative and survival stimuli. The two receptors were co-characterized in the ovary, where they modulate different intracellular signaling cascades, according to the expression level and developmental stage of ovarian follicles. Moreover, they may physically interact to form heteromeric assemblies, suggestive of a new mode of action to regulate FSH-specific signals, and likely determining the follicular fate between atresia and dominance. The knowledge of FSH and estrogen membrane receptors provides a new, deeper level of comprehension of human reproduction.


Subject(s)
Receptors, Estrogen , Receptors, FSH , Female , Humans , Receptors, FSH/genetics , Estrogens , Ovary , Follicle Stimulating Hormone
20.
Fertil Steril ; 120(5): 1061-1070, 2023 11.
Article in English | MEDLINE | ID: mdl-37532169

ABSTRACT

OBJECTIVE: To determine whether TOP5300, a novel oral follicle-stimulating hormone (FSH) receptor (FSHR) allosteric agonist, elicits a different cellular response than recombinant human FSH (rh-FSH) in human granulosa cells from patients undergoing in vitro fertilization. DESIGN: Basic science research with a preclinical allosteric FSHR agonist. SETTING: University hospital. PATIENT(S): Patients with infertility at a single academic fertility clinic were recruited under an Institutional Review Board-approved protocol. Primary granulosa cell cultures were established for 41 patients, of whom 8 had normal ovarian reserve (NOR), 17 were of advanced reproductive age (ARA), 12 had a diagnosis of polycystic ovary syndrome (PCOS), and 4 had a combination of diagnoses, such as ARA and PCOS. INTERVENTION(S): Primary granulosa-lutein (GL) cell cultures were treated with rh-FSH, TOP5300, or vehicle. MAIN OUTCOME MEASURE(S): Estradiol (E2) production using enzyme-linked immunosorbent assay, steroid pathway gene expression of StAR and aromatase using quantitative polymerase chain reaction, and FSHR membrane localization using immunofluorescence were measured in human GL cells. RESULT(S): TOP5300 consistently stimulated E2 production among patients with NOR, ARA, and PCOS. Recombinant FSH was the more potent ligand in GL cells from patients with NOR but was ineffective in cells from patients with ARA or PCOS. The lowest level of FSHR plasma membrane localization was seen in patients with ARA, although FSHR localization was more abundant in cells from patients with PCOS; the highest levels were present in cells from patients with NOR. The localization of FSHR was not affected by TOP5300 relative to rh-FSH in any patient group. TOP5300 stimulated greater expression of StAR and CYP19A1 across cells from all patients with NOR, ARA, and PCOS combined, although rh-FSH was unable to stimulate StAR and aromatase (CYP19A1) expression in cells from patients with PCOS. TOP5300-induced expression of StAR and CYP19A1 mRNA among patients with ARA and NOR was consistently lower than that observed in cells from patients with PCOS. CONCLUSION(S): TOP5300 appears to stimulate E2 production and steroidogenic gene expression from GL cells more than rh-FSH in PCOS, relative to patients with ARA and NOR. It does not appear that localization of FSHR at cell membranes is a limiting step for TOP5300 or rh-FSH stimulation of steroidogenic gene expression and E2 production.


Subject(s)
Polycystic Ovary Syndrome , Receptors, FSH , Female , Humans , Receptors, FSH/genetics , Receptors, FSH/metabolism , Follicle Stimulating Hormone, Human/pharmacology , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , Aromatase/genetics , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/metabolism , Gonadal Steroid Hormones/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL