Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.277
Filter
1.
F S Sci ; 5(1): 16-23, 2024 Feb.
Article in English | MEDLINE | ID: mdl-39382269

ABSTRACT

OBJECTIVE: To investigate the direct effect of growth differentiation factor 9 (GDF9) on androgen production in human theca cells. DESIGN: Experimental study. SETTING: Tertiary hospital-based research laboratory. PATIENT(S): Women who underwent in vitro fertilization and intracytoplasmic sperm injections at our clinic were included in this study. INTERVENTION(S): Primary cultured human theca cells from women undergoing in vitro fertilization and intracytoplasmic sperm injection treatment were treated with GDF9, an activin receptor-like kinase 5 (ALK5) inhibitor, and a SMAD4 agonist. MAIN OUTCOME MEASURE(S): The expression of androgen synthesis-related genes StAR, CYP17A1, and LHCGR, levels of androstenedione and testosterone, phosphorylation of SMAD2/3, and the interaction between bone morphogenic protein-activated type II receptor and ALK5 were evaluated using reverse transcription-quantitative polymerase chain reaction, Western blot, enzyme-linked immunosorbent assays, and coimmunoprecipitation assays, respectively. RESULT(S): Growth differentiation factor 9 decreased StAR, CYP17A1, and LHCGR expression levels in human theca cells, which was prevented by treatment with the ALK5 inhibitor, and suppressed production of androgen in human theca cells. Growth differentiation factor 9 increased SMAD2/3 phosphorylation, and the ALK5 inhibitor also suppressed this effect. Bone morphogenic protein-activated type II receptor and ALK5 bound to each other after GDF9 stimulation. The SMAD4 agonist kartogenin also decreased messenger RNA levels of StAR and CYP17A1 and protein levels of StAR in human theca cells. CONCLUSION(S): Growth differentiation factor 9 can activate the bone morphogenic protein-activated type II receptor-ALK5-SMAD2/3 signaling pathway, suppress CYP17A1 expression, and decrease androgen production in human theca cells.


Subject(s)
Growth Differentiation Factor 9 , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta , Smad2 Protein , Smad3 Protein , Steroid 17-alpha-Hydroxylase , Theca Cells , Humans , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Theca Cells/metabolism , Theca Cells/drug effects , Female , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/genetics , Growth Differentiation Factor 9/metabolism , Growth Differentiation Factor 9/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Smad2 Protein/metabolism , Smad2 Protein/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Androgens/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , Smad4 Protein/metabolism , Smad4 Protein/genetics , Phosphorylation/drug effects , Cells, Cultured , Oocytes/metabolism , Oocytes/drug effects , Androstenedione/metabolism , Testosterone/metabolism , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction/drug effects
2.
BMC Genomics ; 25(1): 800, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39182029

ABSTRACT

BACKGROUND: The Gα family plays a crucial role in the complex reproductive regulatory network of teleosts. However, the characterization and function of Gα family members, especially Gαq, remain poorly understood in teleosts. To analyze the characterization, expression, and function of grass carp (Ctenopharyngodon idella) Gαq, we identified the Gα family members in grass carp genome, and analyzed the expression, distribution, and signal transduction of Gαq/gnaq. We also explored the role of Gαq in the reproductive regulation of grass carp. RESULTS: Our results showed that the grass carp genome contains 27 Gα genes with 46 isoforms, which are divided into four subfamilies: Gαs, Gαi/o, Gαq/11, and Gα12/13. The expression level of Cignaq in the testis was the highest and significantly higher than in other tissues, followed by the hypothalamus and brain. The luteinizing hormone receptor (LHR) was mainly localized to the nucleus in grass carp oocytes, with signals also present in follicular cells. In contrast, Gαq signal was mainly found in the cytoplasm of oocytes, with no signal in follicular cells. In the testis, Gαq and LHR were co-localized in the cytoplasm. Furthermore, the grass carp Gαq recombinant protein significantly promoted Cipgr expression. CONCLUSIONS: These results provided preliminary evidence for understanding the role of Gαq in the reproductive regulation of teleosts.


Subject(s)
Carps , Reproduction , Animals , Carps/genetics , Carps/metabolism , Reproduction/genetics , Fish Proteins/genetics , Fish Proteins/metabolism , Male , Female , Signal Transduction , Phylogeny , Genome , Testis/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Oocytes/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.
Am J Reprod Immunol ; 91(6): e13886, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39031603

ABSTRACT

PROBLEM: The luteinizing hormone (LH), produced by gonadal and nongonadal cells in the anterior pituitary gland play a critical role in human sexual development and reproduction. It is required for the induction of ovulation in females and sex steroid hormone production in both males and females. It is also an important player in early pregnancy events in oviducts and in absence of LH signalling, the uterus cannot initiate pregnancy. LH works through its receptor LHCGR. Therefore, it is quite important to figure out those mutations that have the potential to affect the structure and function of both LH and LHR. MATERIALS AND METHODS: Various in silico tools were employed in the study for the data mining of SNPs and predicting their possible impact on the structure and function of the protein. ConSurf analysis predicted V454I and I161K are exposed residues in the 2D structure of protein and highly conserved in protein structure. PSIPRED and Swiss Modeller were employed to predict the 2D and 3D structure of mutated receptor protein. FT site server predicted both substitutions were involved in the ligand-binding site RESULTS: By present analysis, we have found that R59G in LHα, Q74R and T78N in LHß and V454I and I161K in LHCGR are the most deleterious nsSNPs affecting the structure and function of the protein. CONCLUSION: These SNPs are still uncharacterised; hence providing a baseline for validation of their association with the susceptibility of diseases and develop personalised therapeutics.


Subject(s)
Computational Biology , Luteinizing Hormone , Polymorphism, Single Nucleotide , Receptors, LH , Female , Humans , Male , Binding Sites , Computational Biology/methods , Computer Simulation , Luteinizing Hormone/metabolism , Models, Molecular , Mutation/genetics , Protein Conformation , Receptors, LH/genetics , Receptors, LH/metabolism
5.
Anim Reprod Sci ; 268: 107534, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39047429

ABSTRACT

The spatiotemporal transcription of follicle-stimulating hormone receptor (FSHR) and luteinizing hormone/human chorionic gonadotropin receptor (LHCGR) are crucial events for follicular development. However, their regulatory mechanisms are unclear. DNA methylation and histone acetylation are the main epigenetic modifications, and play important roles in transcriptional expression, which regulate cell responses including cell proliferation, senescence and apoptosis. This review will discuss the dynamic epigenetic modifications of FSHR and LHCGR that occur during the process of follicular development and their response to gonadotropins. In addition, some alteration patterns that occur during these epigenetic modifications, as well as their retrospect retrotransposons, which regulate the gene expression levels of FSHR and LHCGR will be discussed.


Subject(s)
Epigenesis, Genetic , Animals , Ovarian Follicle/metabolism , Ovarian Follicle/physiology , Female , Humans , Receptors, Gonadotropin/genetics , Receptors, Gonadotropin/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167165, 2024 06.
Article in English | MEDLINE | ID: mdl-38653355

ABSTRACT

The mechanisms that underly reproductive hormone effects on cognition, neuronal plasticity, and AD risk, particularly in relation to gonadotropin LH receptor (LHCGR) signaling, remain poorly understood. To address this gap in knowledge and clarify the impact of circulating steroid hormones on the therapeutic effects of CNS LHCGR activation, we delivered the LHCGR agonist human chorionic gonadotropin (hCG) intracerebroventricularly (ICV) and evaluated functional, structural, plasticity-related signaling cascades, Aß pathology, and transcriptome differences in reproductively intact and ovariectomized (OVX) APP/PS1 AD female mice. Here we demonstrate that CNS hCG delivery restored function to wild-type levels only in OVX APP/PS1 mice. Spine density was increased in all hCG treated groups independently of reproductive status. Notably, increases in BDNF signaling and cognition, were selectively upregulated only in the OVX hCG-treated group. RNA sequencing analyses identified a significant increase in peripheral myeloid and pro-inflammatory genes within the hippocampi of the OVX group that were completely reversed by hCG treatment, identifying a potential mechanism underlying the selective therapeutic effect of LHCGR activation. Interestingly, in intact mice, hCG administration mimicked the effects of gonadectomy. Together, our findings indicate that CNS LHCGR agonism in the post-menopausal context is beneficial through trophic and immune mechanisms. Our findings also underscore the presence of a steroid-LHCGR mechanistic interaction that is unexplored yet potentially meaningful to fully understand "post-menopausal" brain function and CNS hormone treatment response.


Subject(s)
Alzheimer Disease , Chorionic Gonadotropin , Disease Models, Animal , Receptors, LH , Animals , Female , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Mice , Chorionic Gonadotropin/pharmacology , Receptors, LH/metabolism , Receptors, LH/genetics , Receptors, LH/agonists , Mice, Transgenic , Ovariectomy , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Humans , Reproduction/drug effects , Presenilin-1/genetics , Presenilin-1/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Hippocampus/metabolism , Hippocampus/drug effects , Signal Transduction/drug effects , Cognition/drug effects
7.
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
8.
Endocrinology ; 165(6)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38679471

ABSTRACT

The glycoprotein receptors, members of the large G protein-coupled receptor family, are characterized by a large extracellular domains responsible for binding their glycoprotein hormones. Hormone-receptor interactions are traditionally analyzed by ligand-binding assays, most often using radiolabeling but also by thermal shift assays. Despite their high sensitivity, these assays require appropriate laboratory conditions and, often, purified plasma cell membranes, which do not provide information on receptor localization or activity because the assays typically focus on measuring binding only. Here, we apply bioluminescence resonance energy transfer in living cells to determine hormone-receptor interactions between a Gaussia luciferase (Gluc)-luteinizing hormone/chorionic gonadotropin receptor (LHCGR) fusion and its ligands (human chorionic gonadotropin or LH) fused to the enhanced green fluorescent protein. The Gluc-LHCGR, as well as other Gluc-G protein-coupled receptors such as the somatostatin and the C-X-C motif chemokine receptors, is expressed on the plasma membrane, where luminescence activity is equal to membrane receptor expression, and is fully functional. The chimeric enhanced green fluorescent protein-ligands are properly secreted from cells and able to bind and activate the wild-type LHCGR as well as the Gluc-LHCGR. Finally, bioluminescence resonance energy transfer was used to determine the interactions between clinically relevant mutations of the hormones and the LHCGR that show that this bioassay provides a fast and effective, safe, and cost-efficient tool to assist the molecular characterization of mutations in either the receptor or ligand and that it is compatible with downstream cellular assays to determine receptor activation/function.


Subject(s)
Green Fluorescent Proteins , Protein Binding , Humans , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Receptors, LH/metabolism , Receptors, LH/genetics , Luciferases/metabolism , Luciferases/genetics , Animals , Bioluminescence Resonance Energy Transfer Techniques/methods , Chorionic Gonadotropin/metabolism , HEK293 Cells , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Energy Transfer , Glycoproteins/metabolism , Luminescent Measurements/methods
9.
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
10.
Hormones (Athens) ; 23(2): 305-312, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38526829

ABSTRACT

PURPOSE: Leydig cell hypoplasia (LCH) type II is a rare disease with only a few cases reported. Patients presented with hypospadias, micropenis, undescended testes, or infertility. In this study, we report a new patient with compound heterozygous variants in the LHCGR gene and LCH type II phenotype. METHODS: Whole exome sequencing (WES) was performed followed by Sanger sequencing to confirm the detected variants in the patient and his parents. RESULTS: A novel missense variant (p.Phe444Cys) was identified in a highly conserved site and is verified to be in trans with the signal peptide's 33-bases insertion variant. CONCLUSION: Our research provides a more comprehensive clinical and genetic spectrum of Leydig cell hypoplasia type II. It highlighted the importance of WES in the diagnosis of this uncommon genetic disorder as well as the expansion of the genotype of LCH type II.


Subject(s)
Disorder of Sex Development, 46,XY , Phenotype , Receptors, LH , Humans , Male , Receptors, LH/genetics , Disorder of Sex Development, 46,XY/genetics , Disorder of Sex Development, 46,XY/diagnosis , Exome Sequencing , Protein Sorting Signals/genetics , Mutation, Missense , Steroid Metabolism, Inborn Errors/genetics , Alleles , Testis/abnormalities
11.
Endocrine ; 83(2): 473-482, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37828397

ABSTRACT

PURPOSE: Transient pregnancy-induced Cushing's syndrome is a rare condition characterized by the manifestation of symptoms solely during pregnancy, which typically resolve spontaneously following delivery or miscarriage. While it has been established that GNAS is associated with adrenal tumors, its specific role in the pathogenesis of pregnancy-induced Cushing's syndrome remains uncertain.This work aims to examine the association between GNAS mutation and pregnancy-induced Cushing's syndrome. METHODS: DNA was extracted from patients' peripheral blood and tumor tissues for whole-exome sequencing (WES) and Sanger sequencing. We used AlphaFold to predict the protein structure of wild-type and mutant GNAS and to make functional predictions, and immunohistochemistry was used to detect disease-associated protein expression. A review and summary of reported cases of transient pregnancy-induced Cushing's syndrome induced by pregnancy was conducted. RESULTS: Using WES, we identified a somatic mutation in GNAS (NM_000516, c.C601T, p.R201C) that was predicted to have a deleterious effect using computational methods, such as AlphaFold. Human chorionic gonadotropin (hCG) stimulation tests had weakly positive results, and immunohistochemical staining of adrenal adenoma tissue also revealed positivity for luteinizing hormone/chorionic gonadotropin receptor (LHCGR) and cytochrome P450 family 11 subfamily B member 1 (CYP11B1). We reviewed 15 published cases of transient Cushing's syndrome induced by pregnancy. Among these cases, immunohistochemical staining of the adrenal gland showed positive LHCGR expression in 3 case reports, similar to our findings. CONCLUSION: Transient pregnancy-induced Cushing's syndrome may be associated with somatic GNAS mutations and altered adrenal pathology due to abnormal activation of LHCGR.


Subject(s)
Cushing Syndrome , Female , Pregnancy , Humans , Cushing Syndrome/diagnosis , Receptors, LH/genetics , Receptors, LH/metabolism , Luteinizing Hormone/metabolism , Chorionic Gonadotropin , Mutation , Hydrocortisone , Chromogranins/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics
12.
J Pak Med Assoc ; 73(12): 2458-2461, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38083932

ABSTRACT

A male child, aged seven months, visited the out patients clinic of the National Institute of Child Health, Karachi, in May 2020 with the features of iso-sexual puberty. After ruling out the more common causes of early puberty, like congenital adrenal hyperplasia and tumours secreting chorionic gonadotropin hormone, hormonal assessment indicated raised testosterone independent of gonadotropin. The volume of the testicles was symmetric and testicular ultrasonography revealed no mass. Genetic analysis for the LHCGR gene was performed for confirmation which revealed activating heterozygous missense pathogenic mutation in c.1732G>T (p.Asp578Tyr). This is the first reported case of testotoxicosis (FMPP) from Pakistan which was genetically confirmed.


Subject(s)
Puberty, Precocious , Child , Humans , Infant , Male , Chorionic Gonadotropin , Mutation , Mutation, Missense , Pakistan , Puberty, Precocious/genetics , Receptors, LH/genetics
13.
J Ovarian Res ; 16(1): 189, 2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37691102

ABSTRACT

BACKGROUND: The concentration of human chorionic gonadotropin (hCG)/ luteinizing hormone (LH) after triggering is generally accepted as a predictor of the normal ovarian response to the trigger, but few studies have explored the distribution model of concentration and its impact on oocyte yield. Genetic variations in LHCGR, known as a receptor for hCG and LH, also play a role in oocyte maturation and retrieval. The objective of the study was to investigate the impact of concentrations of hCG/LH after triggering on oocyte yield and its association with genetic variants of LHCGR. METHODS: A retrospective cohort study including 372 antagonist IVF cycles, in which 205 received the recombinant hCG trigger and 167 received the gonadotropin-releasing hormone agonist (GnRH-a) trigger, was conducted. The post-trigger concentrations of hCG/LH and the LHCGR N312S (rs2293275) genotype were evaluated in patients to analyse the impact of these factors on oocyte yield. RESULTS: The oocyte retrieval rate (ORR) increased significantly among the low-, medium- and high-hCG-concentration groups (0.91 ± 0.25, 0.99 ± 0.23 and 1.08 ± 0.19, P < 0.001) and among the low-, medium- and high-LH-concentration groups (0.80 ± 0.29, 0.95 ± 0.21 and 1.07 ± 0.19, P < 0.001). The Pearson correlation coefficient between the post-trigger hCG concentration and ORR was 0.242 (P < 0.001), and that between the LH concentration and ORR was 0.454 (P < 0.001). After adjustment for confounding factors, high post-trigger LH concentrations remained associated with the significantly higher ORRs (adjusted R2 = 0.541, P < 0.001). Patients with the AG genotype of LHCGR N312S were more likely to have low post-trigger LH concentrations (46.10 IU/L versus 60.91 IU/L, P < 0.001) and a significantly lower ORR (0.85 versus 0.96, P = 0.042) than patients with the GG genotype after the GnRH-a trigger. CONCLUSIONS: The post-trigger LH concentration can positively predict oocyte yield in antagonist IVF cycles, and patients with the AG genotype of LHCGR rs2293275 could have a suboptimal oocyte yield using the GnRH-a trigger.


Subject(s)
Luteinizing Hormone , Oocytes , Receptors, LH , Humans , Chorionic Gonadotropin , Gonadotropin-Releasing Hormone/genetics , Receptors, G-Protein-Coupled , Retrospective Studies , Receptors, LH/genetics
14.
Adv Sci (Weinh) ; 10(29): e2300993, 2023 10.
Article in English | MEDLINE | ID: mdl-37697644

ABSTRACT

Hereditary primary hypogonadism (HPH), caused by gene mutation related to testosterone synthesis in Leydig cells, usually impairs male sexual development and spermatogenesis. Genetically corrected stem Leydig cells (SLCs) transplantation may provide a new approach for treating HPH. Here, a novel nonsense-point-mutation mouse model (LhcgrW495X ) is first generated based on a gene mutation relative to HPH patients. To verify the efficacy and feasibility of SLCs transplantation in treating HPH, wild-type SLCs are transplanted into LhcgrW495X mice, in which SLCs obviously rescue HPH phenotypes. Through comparing several editing strategies, optimized PE2 protein (PEmax) system is identified as an efficient and precise approach to correct the pathogenic point mutation in Lhcgr. Furthermore, delivering intein-split PEmax system via lentivirus successfully corrects the mutation in SLCs from LhcgrW495X mice ex vivo. Gene-corrected SLCs from LhcgrW495X mice exert ability to differentiate into functional Leydig cells in vitro. Notably, the transplantation of gene-corrected SLCs effectively regenerates Leydig cells, recovers testosterone production, restarts sexual development, rescues spermatogenesis, and produces fertile offspring in LhcgrW495X mice. Altogether, these results suggest that PE-based gene editing in SLCs ex vivo is a promising strategy for HPH therapy and is potentially leveraged to address more hereditary diseases in reproductive system.


Subject(s)
Hypogonadism , Leydig Cells , Receptors, LH , Animals , Humans , Male , Mice , Cell Differentiation , Hypogonadism/genetics , Hypogonadism/therapy , Leydig Cells/transplantation , Mutation , Receptors, G-Protein-Coupled , Testosterone/metabolism , Receptors, LH/genetics
15.
Int J Mol Sci ; 24(15)2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37569429

ABSTRACT

We demonstrate here that highly sensitive in vitro bioassays for FSH, TSH, and PTH can be set up in mouse Leydig Tumor Cells (mLTC), in addition to the normal LH/CG bioassay, after they were transfected with expression vectors encoding the corresponding Gs Protein-Coupled Receptors (GsPCR), such as FSHR, TSHR, or PTHR. Although the ß2 adrenergic receptor is also a GsPCR, its expression in mLTC led to a significant but very low cAMP response compared to those observed with FSH, TSH, or PTH. Similarly, after transfection of the GiPCR MT1 melatonin receptor, we did not observe any inhibitory effect by melatonin of the LH or hCG stimulation. Interestingly, after transfection of mLTC with the human kisspeptin receptor (hKpR), which is a GqPCR, we observed a dose-dependent synergy of 10-12-10-7 M kisspeptin variants with a fixed concentration of 0.3 nM LH or hCG. Without any exogenous receptor transfection, a 2 h preincubation with OT or AVP led to a dose-dependent cAMP response to a fixed dose of LH or hCG. Therefore, highly sensitive in vitro bioassays for various hormones and other GPCR ligands can be set up in mLTC to measure circulating concentrations in only 3-10 µL of blood or other body fluids. Nevertheless, the development of an LHRKO mLTC cell line will be mandatory to obtain strict specificity for these bioassays to eliminate potential cross-reaction with LH or CG.


Subject(s)
Kisspeptins , Receptors, LH , Mice , Animals , Humans , Receptors, LH/genetics , Receptors, LH/metabolism , Kisspeptins/metabolism , Ligands , Cyclic AMP/metabolism , Signal Transduction , Receptors, G-Protein-Coupled , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/metabolism , Thyrotropin/metabolism , Chorionic Gonadotropin/metabolism
16.
Res Vet Sci ; 161: 132-137, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37384971

ABSTRACT

Testosterone in male mammals is mainly secreted by testicular Leydig cells, and its secretion process is regulated by the hypothalamic-pituitary-gonadal axis. After receiving the luteinizing hormone (LH) stimulus signal, the lutropin/choriogonadotropin receptor (LHCGR) on the Leydig cell membrane transfers the signal into the cell and finally increases the secretion of testosterone by upregulating the expression of steroid hormone synthase. In previous experiments, we found that interfering with the expression of the Luman protein can significantly increase testosterone secretion in MLTC-1 cells. In this experiment, we found that knockdown of Luman in MLTC-1 cells significantly increased the concentration of cAMP and upregulated the expression of AC and LHCGR. Moreover, an analysis of the activity of the LHCGR promoter by a dual luciferase reporter system showed that knockdown of Luman increased the activity of the LHCGR promoter. Therefore, we believe that knockdown of Luman increased the activity of the LHCGR promoter and upregulated the expression of LHCGR, thereby increasing the concentration of intracellular cAMP and ultimately leading to an increase of testosterone secretion by MLTC-1 cells.


Subject(s)
Leydig Cells , Receptors, LH , Male , Animals , Receptors, LH/genetics , Receptors, LH/metabolism , Testosterone/metabolism , Testis/metabolism , Luteinizing Hormone/pharmacology , Luteinizing Hormone/metabolism , Mammals
17.
J Endocrinol ; 257(1)2023 04 01.
Article in English | MEDLINE | ID: mdl-36705256

ABSTRACT

There are many previous reports on the effects of ethanol on physiological function, including reports of elevated blood estrogen levels in women who drank alcohol. However, the mechanism of ethanol's effects on ovarian functions, such as follicle development and hormone secretion, has not been fully clarified. Therefore, in this study, we investigated the impacts of ethanol on these phenomena and their mechanisms using a primary culture system of rat ovarian granulosa cells (GCs). In the present experiment, groups were created in which follicle-stimulating hormone (FSH) or ethanol was added alone or FSH and ethanol were co-added, and mRNA and protein expression in each group was measured for luteinizing hormone receptor (LHR) and sex steroid hormone synthase, as well as for estradiol (E2) production, cAMP production, and FSH receptor (FSHR) internalization rate. The addition of FSH induced mRNA expression of LHR and aromatase, which led to membrane LHR expression and E2 production. The coexistence of ethanol enhanced all these responses. The action of FSH is exerted via cAMP, and the co-addition of ethanol enhanced this cAMP production. Ethanol alone did not induce cAMP production. The enhancing effect of ethanol was also observed for cAMP induced by cholera toxin. Ethanol had no significant effect on the internalization rate of FSHR. In conclusion, ethanol increased FSH-stimulated cAMP production by increasing the activity of adenylyl cyclase, which enhanced FSH actions in rat GCs. Alcohol is an exacerbating factor in several female hormone-related diseases, and the mechanism of ethanol-induced increase in estrogen secretion revealed in this study may be involved in the pathogenesis of these diseases.


Subject(s)
Ethanol , Follicle Stimulating Hormone , Rats , Female , Animals , Ethanol/pharmacology , Ethanol/metabolism , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/metabolism , Granulosa Cells/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , RNA, Messenger/metabolism , Cells, Cultured , Estrogens/pharmacology , Estrogens/metabolism
18.
Biochem Genet ; 61(4): 1418-1432, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36633772

ABSTRACT

 Polycystic ovary syndrome (PCOS) is a common multifaceted endocrine disorder among reproductive-aged women. Deranged luteinizing hormone levels and associated downstream signaling cascade mediated by its receptor luteinizing hormone chorionic gonadotropin receptor (LHCGR) are pivotal in the etiopathogenesis of PCOS. Genetic variations in the LHCGR have been associated with PCOS risk. However, the results are mixed and inconclusive. We evaluated the association of the LHCGR rs2293275 polymorphic variant with PCOS risk and its association with clinico-biochemical features of PCOS. 120 confirmed PCOS cases and an equal number of age-matched controls were subjected to clinical, biochemical, and hormonal investigations. Genotyping for rs2293275 was performed using polymerase chain reaction-restriction fragment length polymorphism. Logistic regression models were used to calculate odds ratios (ORs) at 95% confidence intervals (95% CIs). In the current study, PCOS cases reported a lower number of menstrual cycles per year than respective controls. A significantly higher BMI, Ferriman Galway score, levels of serum testosterone, insulin, TSH, FSH, and fasting glucose were observed in cases than in controls (p < 0.01). Compared to GG carriers, we observed a higher risk of developing PCOS in the subjects who harbored GA (OR 10.4, p < 0.0001) or AA (OR 7.73, p = 0.02) genotype. The risk persisted in the dominant model (GA + AA) as well (OR 10.29, p = 0.01). On stratification, a higher risk of developing PCOS was observed in variant genotype carriers who had a family history of either type two diabetes mellitus (OR 117; p < 0.0001) or hirsutism (OR 79; p < 0.0001). We also found significantly elevated levels of serum LH levels in the subject harboring GA and AA genotypes when compared to GG carriers. In the present study, we report a significant association of the LHCGR rs2293275 variant with the PCOS risk.


Subject(s)
Polycystic Ovary Syndrome , Receptors, LH , Humans , Female , Adult , Receptors, LH/genetics , Polycystic Ovary Syndrome/genetics , Genetic Predisposition to Disease , Case-Control Studies , Gene Frequency , Luteinizing Hormone/genetics , Polymorphism, Single Nucleotide
19.
Biol Reprod ; 108(2): 258-268, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36214501

ABSTRACT

Angiogenesis within the ovarian follicle is an important component of ovulation. New capillary growth is initiated by the ovulatory surge of luteinizing hormone (LH), and angiogenesis is well underway at the time of follicle rupture. LH-stimulated follicular production of vascular growth factors has been shown to promote new capillary formation in the ovulatory follicle. The possibility that LH acts directly on ovarian endothelial cells to promote ovulatory angiogenesis has not been addressed. For these studies, ovaries containing ovulatory follicles were obtained from cynomolgus macaques and used for histological examination of ovarian vascular endothelial cells, and monkey ovarian microvascular endothelial cells (mOMECs) were enriched from ovulatory follicles for in vitro studies. mOMECs expressed LHCGR mRNA and protein, and immunostaining confirmed LHCGR protein in endothelial cells of ovulatory follicles in vivo. Human chorionic gonadotropin (hCG), a ligand for LHCGR, increased mOMEC proliferation, migration and capillary-like sprout formation in vitro. Treatment of mOMECs with hCG increased cAMP, a common intracellular signal generated by LHCGR activation. The cAMP analog dibutyryl cAMP increased mOMEC proliferation in the absence of hCG. Both the protein kinase A (PKA) inhibitor H89 and the phospholipase C (PLC) inhibitor U73122 blocked hCG-stimulated mOMEC proliferation, suggesting that multiple G-proteins may mediate LHCGR action. Human ovarian microvascular endothelial cells (hOMECs) enriched from ovarian aspirates obtained from healthy oocyte donors also expressed LHCGR. hOMECs also migrated and proliferated in response to hCG. Overall, these findings indicate that the LH surge may directly activate ovarian endothelial cells to stimulate angiogenesis of the ovulatory follicle.


Subject(s)
Endothelial Cells , Neovascularization, Physiologic , Ovary , Receptors, LH , Animals , Female , Humans , Chorionic Gonadotropin/pharmacology , Chorionic Gonadotropin/metabolism , Endothelial Cells/metabolism , Luteinizing Hormone/pharmacology , Luteinizing Hormone/metabolism , Macaca fascicularis , Neovascularization, Physiologic/physiology , Ovarian Follicle/metabolism , Ovary/blood supply , Ovary/metabolism , Ovulation/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism
20.
Anim Biotechnol ; 34(7): 2285-2294, 2023 Dec.
Article in English | MEDLINE | ID: mdl-35714982

ABSTRACT

To evaluate the efficacy of oral immunization with active kisspeptin DNA vaccine on the expression of hormone receptor mRNA. For this study, ten 56-day-old Hu breed ram lambs were randomly assigned to the treatment and control groups (n = 5). Treatment Experimental group received C500/pKS-asd and the control group received C500/pVAX-asd (aspartate-ß semialdehyde dehydrogenase orally on days 0, 28, and 56, and blood samples were taken at each immunization interval (14-day) and tissues samples were collected at the end of the experimental period (day 98). The collected samples were stored in the refrigerator at -20 °C and liquid nitrogen, respectively, for laboratory examination. Total RNA was extracted from samples using TRIzol reagent and quantitative real-time polymerase chain reaction (QPCR) was used to quantify the levels of KISS1, G protein-coupled receptor-54 (Kiss1r), and gonadotrophin-releasing hormone (GnRH) mRNA in the hypothalamus. Levels of luteinizing hormone receptor (LHR) and luteinizing hormone beta (LHß) mRNA, and follicle-stimulating hormone receptor (FSHR) and follicle-stimulating hormone beta (FSHß) mRNA in the testes and pituitary were analyzed, respectively. Further, gonadotropin-releasing hormone receptor (GnRHR) mRNA expression level in the pituitary was measured. Moreover, the Kiss1r concentration level in the blood was measured using an indirect ELISA. The concentration of Kiss1r in the blood was lower in the treatment group than in the control group (p < 0.05). The levels of testicular FSHR and LHR mRNA were significantly lower in the treatment group (p < 0.05) when compared to the control group. Furthermore, the treatment group's levels of hypothalamic KISS1, Kiss1r, and GnRH mRNA were significantly lower (p < 0.05) than the controls. LH, FSH, and GnRHR mRNA expression in the pituitary were also significantly lower in the treatment group (p < 0.01 and p < 0.05, respectively). These findings imply that oral immunization with active kisspeptin DNA vaccine suppresses hormone receptor mRNA expression in the ram lambs.


Subject(s)
Kisspeptins , Vaccines, DNA , Sheep/genetics , Animals , Male , Kisspeptins/genetics , Receptors, Kisspeptin-1 , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Sheep, Domestic/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, LH/genetics , Immunization/veterinary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Follicle Stimulating Hormone/genetics
SELECTION OF CITATIONS
SEARCH DETAIL