Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Assist Reprod Genet ; 41(5): 1387-1401, 2024 May.
Article in English | MEDLINE | ID: mdl-38656738

ABSTRACT

OBJECTIVE: Women who are of reproductive age can suffer from polycystic ovary syndrome (PCOS), an endocrine disorder. Anovulatory infertility is mostly caused by aberrant follicular development, which is seen in PCOS patients. Due to the dysfunction of reproductive and endocrine function in PCOS patients, assisted reproduction treatment is one of the main means to obtain clinical pregnancy for PCOS patients. Long non-coding RNA (lncRNA) as a group of functional RNA molecules have been found to participate in the regulation of oocyte function, hormone metabolism, and proliferation and apoptosis of granulosa cells. In this study, we investigated the role of lncRNAs in follicular fluid-derived exosomes and the underlying mechanism of lncRNA LIPE-AS1. METHODS: We used RNA sequencing to analyze the lncRNA profiles of follicular fluid-derived exosomes in PCOS patients and controls. RT-qPCR was performed to detect the expression levels of these lncRNAs in control (n = 30) and PCOS (n = 30) FF exosome samples. Furthermore, we validated the performance of lncRNA LIPE-AS1 in oocyte maturation by in vitro maturation (IVM) experiments in mouse and steroid metabolism in granulosa cells. RESULTS: We found 501 lncRNAs were exclusively expressed in the control group and another 273 lncRNAs were found to be specifically expressed in the PCOS group. LncRNA LIPE-AS1, highly expressed in PCOS exosomes, was related to a poor oocyte maturation and embryo development in PCOS patients. Reduced number of MII oocytes were observed in the LIPE-AS1 group by in vitro maturation (IVM) experiments in mouse. LIPE-AS1 was also shown to modulate steroid metabolism and granulosa cell proliferation and apoptosis by LIPE-AS1/miR-4306/LHCGR axis. CONCLUSION: These findings suggested that the increased expression of LIPE-AS1, facilitated by follicular fluid exosomes, had a significant impact on both oocyte maturation and embryo development. We demonstrated the ceRNA mechanism involving LIPE-AS1, miR-4306, and LHCGR as a regulator of hormone production and metabolism. These findings indicate that LIPE-AS1 is essential in PCOS oocyte maturation and revealed a ceRNA network of LIPE-AS1 and provided new information on abnormal steroid metabolism and oocyte development in PCOS.


Subject(s)
Exosomes , Follicular Fluid , Granulosa Cells , Oocytes , Polycystic Ovary Syndrome , RNA, Long Noncoding , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/pathology , Polycystic Ovary Syndrome/metabolism , Female , Follicular Fluid/metabolism , RNA, Long Noncoding/genetics , Granulosa Cells/metabolism , Granulosa Cells/pathology , Humans , Exosomes/genetics , Exosomes/metabolism , Oocytes/metabolism , Oocytes/growth & development , Mice , Animals , In Vitro Oocyte Maturation Techniques , Adult , Steroids/metabolism , Oogenesis/genetics , Apoptosis/genetics , Cell Proliferation/genetics
2.
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
3.
Anim Biotechnol ; 34(5): 1719-1726, 2023 Nov.
Article in English | MEDLINE | ID: mdl-35297729

ABSTRACT

Single nucleotide polymorphism (SNP) analysis of fertility genes will be useful in improving the genetic potential of dairy cows. A population of 142 cross-bred dairy cows was screened for SNPs in bovine luteinizing hormone choriogonadotropin receptor (LHCGR) and bovine follicle stimulating hormone receptor (FSHR) genes. The effect of reported SNPs on selected fertility traits (average calving interval, average number of services per conception and age at first calving) and milk yield was determined. Altogether six SNPs were detected in animals screened under the present study. Among them, four SNPs (rs41256848, rs41256850, rs465790244, and rs45463781) were located in the exon 11 region of the LHCGR gene and two SNPs (rs43676359 and G-278-A (GU253337) were located in the five upstream region of the FSHR gene. Minor alleles identified for SNPs in the FSHR gene in the studied small population of cows differed from those reported in other research. In this study, only rs45463781 SNP at exon 11 of the LHCGR gene significantly affected the average milk yield in cross-bred cows. The nucleotide change from "G" to "A" negatively affected the average milk yield. Further investigations are needed to confirm the reported association with a larger sample size.


Subject(s)
Fertility , Milk , Female , Cattle/genetics , Animals , Sri Lanka , Genotype , Fertility/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, G-Protein-Coupled/genetics , Lactation/genetics
4.
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
5.
J Obstet Gynaecol Res ; 49(10): 2436-2445, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37462066

ABSTRACT

AIM: The genetic basis of empty follicle syndrome (EFS) is largely unknown, and the aim of this study was to investigate the genetic causes of EFS in primary infertile women. METHODS: Four affected women diagnosed with anovulation were recruited, and whole exome sequencing (WES) was requested for the genetic diagnosis of the cases. One hundred healthy controls were verified by Sanger sequencing. RESULTS: A novel homozygous variant of the LHCGR gene (NM_000233:c.1847C>A) was revealed in one affected individual by WES. Trios analysis of the mutation revealed an autosomal recessive pattern. This LHCGR variant was absent in 100 healthy controls and predicted to be highly damaging to the function of LHCGR. CONCLUSIONS: The novel variant extends the mutational spectrum of the LHCGR gene associated with female sterility, which promotes the prognostic value of testing for LHCGR mutations in infertile women with EFS.


Subject(s)
Infertility, Female , Ovarian Diseases , Humans , Female , Infertility, Female/genetics , Mutation, Missense , Exome Sequencing , Mutation
6.
Int J Mol Sci ; 24(16)2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37628741

ABSTRACT

The ovary is a highly susceptible organ to senescence, and granulosa cells (GCs) have a crucial role in oocyte development promotion and overall ovarian function maintenance. As age advances, GCs apoptosis and dysfunction escalate, leading to ovarian aging. However, the molecular mechanisms underpinning ovarian aging remain poorly understood. In this study, we observed a correlation between the age-related decline of fertility and elevated expression levels of miR-143-3p in female mice. Moreover, miR-143-3p was highly expressed in senescent ovarian GCs. The overexpression of miR-143-3p in GCs not only hindered their proliferation and induced senescence-associated secretory phenotype (SASP) but also impeded steroid hormone synthesis by targeting ubiquitin-conjugating enzyme E2 E3 (Ube2e3) and luteinizing hormone and human chorionic gonadotropin receptor (Lhcgr). These findings suggest that miR-143-3p plays a substantial role in senescence and steroid hormone synthesis in GCs, indicating its potential as a therapeutic target for interventions in the ovarian aging process.


Subject(s)
Estradiol , MicroRNAs , Humans , Female , Animals , Mice , Ovary , Receptors, G-Protein-Coupled , Granulosa Cells , Senescence-Associated Secretory Phenotype , MicroRNAs/genetics
7.
Biochem Biophys Res Commun ; 617(Pt 2): 41-47, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35689841

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide, with the incidence in men being about twice as compared to women. Gender differences may provide clues for finding key targets that mediate the death of dopaminergic (DA) neurons in PD. Luteinizing hormone (LH), analog of human chorionic gonadotropin (hCG), and their receptor, luteinizing hormone/choriogonadotropin receptor (LHCGR), are associated with the pathogenesis of PD. Movement-related symptoms are partially improved by hCG in PD patients. However, the relationship between hCG and PD, as well as its roles in mediating DA neuronal death, has not been elucidated. In this study, we investigated the potential of hCG as a treatment during PD progression. After establishment of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse models, we found that hCG restored the decrease of LHCGR activity caused by down-regulation of LH in the substantia nigra. Furthermore, the reduction of LHCGR activity led to DA neuronal death through knocking down the LHCGR in DA neurons by AAV-mTH-shRNA. Treatment with hCG alleviated the DA neuronal death induced by MPTP. Finally, hCG exerted neuroprotective effects by inhibiting the activation of glycogen synthase kinase 3 beta (GSK3ß) in our MPTP-induced PD mouse and MPP+-treated SH-SY5Y cell models. Together, these results demonstrate that hCG exerts neuroprotective effects for PD through LHCGR, and the inhibition of GSK3ß activation is involved in this protective effect, suggesting that hCG can be taken as a potential therapeutic for the treatment of PD.


Subject(s)
Neuroblastoma , Neurodegenerative Diseases , Neuroprotective Agents , Parkinson Disease , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Chorionic Gonadotropin/pharmacology , Disease Models, Animal , Dopaminergic Neurons/pathology , Female , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Inbred C57BL , Neuroblastoma/pathology , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Substantia Nigra/pathology
8.
BMC Endocr Disord ; 22(1): 335, 2022 Dec 30.
Article in English | MEDLINE | ID: mdl-36585675

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) is an endocrine-metabolic disorder that affects women at their child bearing age. The exact etiology is uncertain, however the involvement of multiple genes and environmental interactions has been proposed for the advancement of PCOS. The aim of present study was to evaluate the association of LHCGR variants (rs2293275 and rs12470652) with PCOS in Punjab. METHODS: The present case-control study comprised a total of 743 women (421 PCOS cases and 322 healthy controls). Genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism technique (PCR-RFLP). Biochemical analysis was carried out to measure the levels of cholesterol, High-density lipoprotein (HDL), Low-density lipoprotein (LDL), Very low-density lipoprotein (VLDL), triglycerides, testosterone, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). All the statistical analysis was done using SPSS (version21, IBM SPSS, NY, USA). RESULTS: The mutant genotype (AA) and mutant allele (A) of rs2293275 conferred 1.7 and 1.3 fold risk, respectively and mutant allele (C) of rs12470652 conferred 2.3 fold risks towards PCOS progression. Levels of cholesterol and triglycerides were elevated and HDL levels were lower in PCOS cases as compared to controls. Total testosterone and luteinizing hormone levels were also found to be higher in PCOS cases. CONCLUSION: Our study postulated that LHCGR variants are playing a cardinal role in the progression of PCOS and can be used to assess the risk of PCOS in women of reproductive age.


Subject(s)
Polycystic Ovary Syndrome , Receptors, LH , Female , Humans , Case-Control Studies , Cholesterol , Genetic Predisposition to Disease , Lipoproteins, LDL , Luteinizing Hormone , Polycystic Ovary Syndrome/genetics , Polymorphism, Single Nucleotide , Receptors, G-Protein-Coupled/genetics , Receptors, LH/genetics , Testosterone , Triglycerides
9.
Reprod Biomed Online ; 43(3): 561-569, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34391684

ABSTRACT

RESEARCH QUESTION: Are there any associations between the variants of FSHR c.2039 G>A (p. Ser680Asn, rs6166) and LHCGR c.935A>G (p. Asn312Ser, rs2293275) and ovarian reserve, ovarian response, clinical pregnancy rate and POSEIDON group? DESIGN: A total of 210 infertile women were enrolled in this prospective study. The gene variants were analysed by the Sanger method. The clinical parameters were analysed based on genotypes. RESULTS: The frequency of heterozygous and homozygous G allele for FSHR c.2039 G>A in the low prognosis group was significantly higher than that in other response groups (P = 0.034); there was no significant association between LHCGR c.935 A>G and ovarian response. Moreover, the serum anti-Müllerian hormone (AMH) concentration, antral follicle count (AFC), oocytes retrieved, metaphase II (MII) oocytes and two-pronuclear (2PN) oocytes in patients with AG genotype for FSHR c.2039 G>A were significantly lower than those with AA genotype. The serum LH concentrations and clinical pregnancy rate of fresh embryo transfer in patients with GG genotype for LHCGR c.935 A>G were significantly higher than that of the AG genotype. In POSEIDON analysis, the low prognosis women with AA genotype for FSHR c.2039 G>A were more likely to appear in subgroup 1 (P = 0.038). CONCLUSION: The FSHR c.2039 G>A variant has a significant beneficial influence on ovarian reserve and ovarian response. The LHCGR c.935 A>G variant is associated with increased clinical pregnancy rate of fresh embryo transfer in infertile women. In addition, the low prognosis women with AA genotype for FSHR c.2039 G>A tend to show better ovarian reserve and prognosis.


Subject(s)
Ovarian Reserve/genetics , Pregnancy Rate , Receptors, FSH/genetics , Receptors, LH/genetics , Adult , Anti-Mullerian Hormone/blood , China/epidemiology , Female , Fertilization in Vitro , Gene Frequency , Genetic Association Studies , Genotype , Humans , Infertility, Female/blood , Infertility, Female/epidemiology , Infertility, Female/genetics , Infertility, Female/therapy , Ovulation Induction , Polymorphism, Single Nucleotide , Pregnancy , Treatment Outcome
10.
BMC Endocr Disord ; 20(1): 62, 2020 May 11.
Article in English | MEDLINE | ID: mdl-32393232

ABSTRACT

BACKGROUND: Pregnancy-induced Cushing's syndrome (CS) with an adrenocortical adenoma overexpressing luteinizing hormone (LH)/human choriogonadotropin (hCG) receptors (LHCGR) has been rarely reported in the literatures. This peculiar condition challenges the canonical diagnosis and management of CS. CASE PRESENTATION: A 27-year-old woman (G2P0A1) presented at 20 weeks gestational age (GA) with overt Cushingoid clinical features. Adrenocorticotropic hormone (ACTH)-independent CS was diagnosed based on undetectable ACTH and unsuppressed cortisol levels by dexamethasone. Magnetic resonance imaging (MRI) scanning without contrast revealed a left adrenal nodule while pituitary MRI scanning was normal. A conservative treatment strategy of controlling Cushingoid comorbidities was conducted. At 36 weeks GA, a caesarean operation was performed and a live female infant was delivered. At 8 weeks after parturition, our patient achieved normalization of blood pressure, blood glucose, serum potassium, and urinary cortisol level spontaneously. During non-pregnancy period, stimulation testing with exogenous hCG significantly evoked a cortisol increase. The woman underwent resection of the adrenal tumor at 6 months after parturition. Immunohistochemistry (IHC) showed the tumor tissue that stained positive for luteinizing hormone (LH)/human choriogonadotropin (hCG) receptor (LHCGR), whereas negative for both melanocortin 2 receptor (MC2R) and G protein-coupled receptor-1 (GPER-1). CONCLUSIONS: Stimulation test with exogenous hCG after parturition is necessary for the diagnosis of pregnancy-induced CS. LHCGR plays an essential role in the pathogenesis of this rare condition.


Subject(s)
Adrenal Cortex Neoplasms/diagnostic imaging , Adrenocortical Adenoma/diagnostic imaging , Cushing Syndrome/diagnosis , Pregnancy Complications, Neoplastic/diagnostic imaging , Receptors, LH/metabolism , Adrenal Cortex Neoplasms/complications , Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex Neoplasms/surgery , Adrenocortical Adenoma/complications , Adrenocortical Adenoma/metabolism , Adrenocortical Adenoma/surgery , Adult , Cushing Syndrome/etiology , Cushing Syndrome/metabolism , Diagnostic Techniques, Endocrine , Female , Humans , Magnetic Resonance Imaging , Pituitary Gland/diagnostic imaging , Pregnancy , Pregnancy Complications/diagnosis , Pregnancy Complications/etiology , Pregnancy Complications/metabolism , Pregnancy Complications, Neoplastic/metabolism , Pregnancy Complications, Neoplastic/surgery
11.
Gynecol Endocrinol ; 36(12): 1136-1139, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32654531

ABSTRACT

INTRODUCTION: Leydig cell hypoplasia (LCH) is an autosomal recessive disease that causes 46, XY sex development disorder. The patients with LCH are usually in the female phenotype and are presented with the complaints of no breast development and primary amenorrhea. In this article, the cases of three siblings who presented with primary amenorrhea and who had LCH were presented. CASE: A 16-year-old patient with female phenotype is presented with primary amenorrhea. Breast development was at Tanner stage 1, the external genitalia were completely in female phenotype. The karyotype was determined as 46, XY. The hormonal analyses revealed that the testosterone synthesis was insufficient despite the high level of luteinizing hormone (LH). Cortisol, ACTH, 17-Hydroxyprogesterone, and AMH levels were normal. LCH diagnosis was considered in the patient with elevated LH and no testosterone synthesis. A new mutation of homozygous c.161 + 4A > G was detected in LHCGR gene. The same mutation was detected in the patient's two siblings with female phenotype and 46, XY karyotype. CONCLUSION: In patients presenting with primary amenorrhea and karyotype 46, XY, there is no testosterone synthesis and if there is LH elevation, LCH should be considered. We found a novel variant in the LHCGR gene in three siblings with karyotype 46, XY and female phenotype.


Subject(s)
Disorder of Sex Development, 46,XY/genetics , Receptors, LH/genetics , Testis/abnormalities , Adolescent , Disorder of Sex Development, 46,XY/blood , Disorder of Sex Development, 46,XY/physiopathology , Female , Homozygote , Humans , Male , Siblings , Testis/physiopathology
12.
J Assist Reprod Genet ; 37(11): 2861-2868, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32860205

ABSTRACT

PURPOSE: To screen novel mutations in LHCGR responsible for empty follicle syndrome and explore the pathological mechanism of mutations. METHODS: Four affected individuals diagnosed with infertility-associated anovulation or oligo-ovulation from three independent families were recruited. Sanger sequencing was used to identify the LHCGR mutations in affected individuals. Western blot was performed to evaluate the effects of mutations on LHCGR protein levels. Immunofluorescence was done to explore the effects of mutations on LHCGR subcellular localization. The ATP levels were measured to infer the functional effects of the mutations on LHCGR. RESULTS: In the present study, three novel biallelic mutations in LHCGR were identified in four affected individuals from three independent families with empty follicle syndrome or oligo-ovulation. All biallelic mutations were inherited from the proband of their parents. The western blot showed that the identified mutations decreased LHCGR protein level and altered the glycosylation pattern. The immunofluorescence showed an ectopic subcellular localization of LHCGR in cultured HeLa cells. Besides, the mutations in LHCGR also reduced the cellular ATP consumption. CONCLUSION: These findings confirm previous studies and expand the mutational spectrum of LHCGR, which will provide genetic diagnostic marker for patients with empty follicle syndrome.


Subject(s)
Infertility, Female/genetics , Polycystic Ovary Syndrome/genetics , Receptors, LH/genetics , Female , HeLa Cells , Humans , Infertility, Female/pathology , Mutation/genetics , Ovarian Follicle/growth & development , Ovarian Follicle/pathology
13.
Int J Mol Sci ; 22(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374698

ABSTRACT

Follicle-stimulating hormone (FSH) and luteinising hormone (LH) play important roles in regulating cell growth and proliferation in the ovary. However, few studies have explored the expression of FSH and LH receptors (FSHR and LHCGR) in ovarian cancer, and their functional roles in cancer progression remain inconclusive. This study investigated the potential impact of both mRNA (FSHR, LHCGR) and protein (FSHR, LHCGR) expression on ovarian cancer progression using publicly available online databases, qRT-PCR (high grade serous ovarian cancers, HGSOC, n = 29 and benign ovarian tumors, n = 17) and immunohistochemistry (HGSOC, n = 144). In addition, we investigated the effect of FSHR and LHCGR siRNA knockdown on the pro-metastatic behavior of serous ovarian cancer cells in vitro. High FSHR or high LHCGR expression in patients with all subtypes of high-grade ovarian cancer was significantly associated with longer progression-free survival (PFS) and overall survival (OS). High FSHR protein expression was associated with increased PFS (p = 0.050) and OS (p = 0.025). HGSOC patients with both high FSHR and high LHCGR protein levels had the best survival outcome, whilst both low FSHR and low LHCGR expression was associated with poorest survival (p = 0.019). Knockdown of FSHR significantly increased the invasion of serous ovarian cancer cells (OVCAR3 and COV362) in vitro. LHCGR knockdown also promoted invasion of COV362 cells. This study highlights that lower FSHR and LHCGR expression is associated with a more aggressive epithelial ovarian cancer phenotype and promotes pro-metastatic behaviour.


Subject(s)
Biomarkers, Tumor/genetics , Neoplasms, Cystic, Mucinous, and Serous/genetics , Ovarian Neoplasms/genetics , Receptors, FSH/genetics , Receptors, LH/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Female , Humans , Middle Aged , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phenotype , Receptors, FSH/metabolism , Receptors, LH/metabolism
14.
Int J Mol Sci ; 22(1)2020 Dec 25.
Article in English | MEDLINE | ID: mdl-33375708

ABSTRACT

(1) The human luteinizing hormone (LH)/chorionic gonadotropin (hCG) receptor (LHCGR) discriminates its two hormone ligands and differs from the murine receptor (Lhr) in amino acid residues potentially involved in qualitative discerning of LH and hCG. The latter gonadotropin is absent in rodents. The aim of the study is to identify LHCGR residues involved in hCG/LH discrimination. (2) Eight LHCGR cDNAs were developed, carrying "murinizing" mutations on aminoacidic residues assumed to interact specifically with LH, hCG, or both. HEK293 cells expressing a mutant or the wild type receptor were treated with LH or hCG and the kinetics of cyclic adenosine monophosphate (cAMP) and phosphorylated extracellular signal-regulated kinases 1/2 (pERK1/2) activation was analyzed by bioluminescence resonance energy transfer (BRET). (3) Mutations falling within the receptor leucine reach repeat 9 and 10 (LRR9 and LRR10; K225S +T226I and R247T), of the large extracellular binding domain, are linked to loss of hormone-specific induced cAMP increase, as well as hCG-specific pERK1/2 activation, leading to a Lhr-like modulation of the LHCGR-mediated intracellular signaling pattern. These results support the hypothesis that LHCGR LRR domain is the interaction site of the hormone ß-L2 loop, which differs between LH and hCG, and might be fundamental for inducing gonadotropin-specific signals. (4) Taken together, these data identify LHCGR key residues likely evolved in the human to discriminate LH/hCG specific binding.


Subject(s)
Amino Acids/chemistry , Binding Sites , Receptors, LH/chemistry , Receptors, LH/metabolism , Amino Acid Sequence , Chorionic Gonadotropin/metabolism , Cyclic AMP/metabolism , HEK293 Cells , Humans , Kinetics , Luteinizing Hormone/metabolism , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Mutation , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Receptors, LH/genetics , Signal Transduction
15.
Gen Comp Endocrinol ; 280: 123-133, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31009604

ABSTRACT

Receptors for follicle-stimulating hormone (Fshr), luteinizing hormone (Lhcgr1 and Lhcgr2) and androgens (Ara and Arb) transduce the hormonal signals that coordinate spermatogenesis, but the factors that regulate the abundance of these transducers in fish testes remain little-understood. To mend this paucity of information, we first determined changes in transcript abundance for these receptors (fshr, lhcgr1, ara and arb) during spermatogenesis induced by human chorionic gonadotropin (hCG) injection in the eel, Anguilla australis. We related our findings to testicular production of the fish androgen, 11-ketotestosterone (11-KT), and to the levels of the transcripts encoding steroidogenic acute regulatory protein (star) and 11ß-hydroxylase (cyp11b), and subsequently evaluated the effects of hCG or 11-KT on mRNA levels of these target genes in vitro. Testicular 11-KT production was greatly increased by hCG treatment, both in vivo and in vitro, and associated with up-regulation of star and cyp11b transcripts. In situ hybridization indicated that testicular fshr mRNA levels were higher in the early stages of hCG-induced spermatogenesis, while lhcgr1 transcripts were most abundant later, once spermatids were observed. In vitro experiments further showed that hCG and its steroidal mediator 11-KT significantly increased fshr transcript abundance. These data provide new angles on the interactions between gonadotropin and androgen signaling during early spermatogenesis. Increases in levels of 11-KT following hCG injection elevated testicular fshr mRNA levels augmenting Fsh sensitivity in the testis. This evidence is suggestive of a positive feedback loop between gonadotropins and 11-KT that may be key to regulating early spermatogenesis in fish.


Subject(s)
Anguilla/genetics , Gene Expression Regulation , Receptors, Androgen/genetics , Receptors, Gonadotropin/genetics , Testis/metabolism , Androgens/metabolism , Anguilla/blood , Animals , Chorionic Gonadotropin/administration & dosage , Chorionic Gonadotropin/pharmacology , Gene Expression Regulation/drug effects , Humans , Male , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, Gonadotropin/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Spermatogenesis/drug effects , Spermatogenesis/genetics , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , Testis/drug effects , Testosterone/analogs & derivatives , Testosterone/blood
16.
Gynecol Endocrinol ; 35(1): 81-85, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30182769

ABSTRACT

To investigate the association between Luteinizing hormone/choriogonadotropin receptor (LHCGR) gene polymorphisms and polycystic ovary syndrome (PCOS). A systematic literature search and meta-analysis using STATA software for included studies. Fourteen case-control studies containing rs13405728, rs4539842, and rs2293275 of LHCGR gene were included, which was comprised of 11,738 PCOS cases and 35,329 controls. Results of the meta-analysis showed a significant association between PCOS and rs13405728 (for G vs. A: OR = 0.735, 95% CI = 0.699-0.773, p<.001; For GG vs. AG + AA: OR = 0.578, 95% CI = 0.436-0.767, p<.001; For GG + AG vs. AA: OR = 0.817, 95% CI = 0.741-0.901, p<.001) in Asian populations, and rs4539842 (for ins/ins vs. ins/non + non/non: OR = 0.686, 95% CI = 0.483-0.974, p=.035) and rs2293275 (for AA vs. AG + GG: OR = 4.115, 95% CI = 1.033-16.38, p=.045) in Caucasian populations, respectively. LHCGR gene variations are population specifically associated with PCOS, which indicated these SNPs in LHCGR may contribute to the pathogenesis of PCOS and could be used as potential biomarkers to predict the risk of PCOS.


Subject(s)
Genetic Predisposition to Disease , Genotype , Polycystic Ovary Syndrome/genetics , Polymorphism, Single Nucleotide , Receptors, LH/genetics , Alleles , Case-Control Studies , Female , Gene Frequency , Humans
17.
Environ Toxicol ; 34(7): 844-852, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30951242

ABSTRACT

Our goals were to investigate whether environmentally relevant doses of T-2 toxin can affect human ovarian granulosa cells' function and to reveal the potential mechanism of T-2 toxin's action. Results showed that T-2 toxin strongly attenuated luteinizing hormone/choriogonadotropin receptor (LHCGR) mRNA expression in follicle-stimulating hormone (FSH)-stimulated human cumulus granulosa cells. Addition of human chorionic gonadotropin was not able to elicit maximal response of ovulatory genes amphiregulin, epiregulin, and progesterone receptor. T-2 toxin reduced mRNA levels of CYP19A1 and steroidogenic acute regulatory protein (STAR) and lowered FSH-stimulated estradiol and progesterone production. Mechanistic experiments demonstrated that T-2 toxin decreased FSH-stimulated cyclic adenosine monophosphate (cAMP) production. Addition of total PDE inhibitor 3-isobutyl-1-methylxanthine prevented T-2 toxin's action on LHCGR, STAR, and CYP19A1 mRNA expression in FSH-stimulated human cumulus granulosa cells. Furthermore, T-2 toxin partially decreased 8-bromoadenosine 3'5'-cyclic monophosphate (8-Br-cAMP)-stimulated LHCGR and STAR, but did not affect 8-Br-cAMP-stimulated CYP19A1 mRNA expression in human cumulus granulosa cells. Overall, our data indicate that environmentally relevant dose of T-2 toxin decreases steroidogenesis and ovulatory potency in human cumulus granulosa cells probably through activation of PDE, thus posing a significant risk for female fertility.


Subject(s)
Aromatase/genetics , Cumulus Cells/drug effects , Cyclic AMP/metabolism , Gonadal Steroid Hormones/biosynthesis , Phosphoproteins/genetics , Receptors, LH/genetics , T-2 Toxin/pharmacology , Adult , Aromatase/metabolism , Cells, Cultured , Chorionic Gonadotropin/metabolism , Cumulus Cells/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Estradiol/metabolism , Female , Follicle Stimulating Hormone/metabolism , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Humans , Phosphoproteins/metabolism , Progesterone/metabolism , RNA, Messenger/metabolism , Receptors, LH/metabolism , Young Adult
18.
Gen Comp Endocrinol ; 263: 21-31, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29660307

ABSTRACT

Follicle-stimulating hormone (Fsh) and luteinizing hormone (Lh), secreted from pituitary, stimulate gonadal function by binding to their cognate receptors FSH receptor (FSHR), and LH/choriogonadotropin receptor (LHCGR). Rohu (Labeo rohita) is a commercially important seasonal breeder freshwater fish species, but till date, the regulation of expression of gonadotropins and their receptors gene during different phases of annual reproductive cycle has not been investigated. We envisaged the critical role of these molecules during seasonal gonadal development in this carp species. We cloned full- length cDNAs of fshra and lhcgrba from rohu testis using RACE (Rapid amplification of cDNA ends) and analyzed their expression along with fsh and lh by quantitative real time PCR (qRT-PCR) assay at various gonadal developmental stages of the annual reproductive cycle. Full-length rohu fshra and lhcgrba cDNA encodes 670 and 716 amino acids respectively, and in adult fish, they were widely expressed in brain, pituitary, gonad, liver, kidney, head kidney, heart, muscle, gill, fin, eye and intestine. In male, both fsh and fshra transcripts showed high level of expression during spermatogenesis, however, in female, expression level was found to be higher in the fully grown oocyte stages. The expression of rohu lh and lhcgrba mRNA increased with increment of gonadosomatic index and showed highest level during spermiation stage in male and fully matured oocyte stage in female. These results together may suggest the involvement of fshra and lhcgrba in regulating function of seasonal gonadal development in rohu.


Subject(s)
Cyprinidae/genetics , Receptors, Gonadotropin/genetics , Animals , Cloning, Molecular , Cyprinidae/metabolism , DNA, Complementary/isolation & purification , DNA, Complementary/metabolism , Female , Gene Expression Profiling/veterinary , Gonads/metabolism , Male , Pituitary Gland/metabolism , Receptors, FSH/metabolism , Receptors, Gonadotropin/isolation & purification , Receptors, Gonadotropin/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Reproduction/genetics , Sequence Analysis, DNA/veterinary , Transcriptome
19.
J Assist Reprod Genet ; 35(9): 1703-1712, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29974367

ABSTRACT

PURPOSE: Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) mediate intracellular functions by binding their specific protein G-coupled gonadotrophin receptor, respectively FSH receptor (FSHR) and LH/choriogonadotrophin receptor (LHCGR). Whereas the expression of FSHR and LHCGR in mammals was considered gonad-specific and cell-specific, studies identified gonadotrophin receptors in human female extragonadal reproductive tissues. This study aims to demonstrate that gonadotrophin receptors are expressed in endometrium and mediates intracellular functions. METHODS: Collected endometria (n = 12) from healthy patients (mean age of 36 ± 6) were primary cultured for 24 h. The presence of gonadotrophin receptors was evaluated by RT-PCR followed by the sequencing of the resulted amplicons and by immunohistochemistry in original samples. Endometrial primary cultures were treated with increasing concentration (range 0-100 ng/ml) of either recombinant human LH (rhLH) or recombinant human FSH (rhFSH). Endometria controls had gonadotrophin replaced by the same volume of the culture medium. In gonadotrophin-treated samples, it was evaluated the intracellular cyclic adenosine monophosphate (cAMP) content by enzymatic immunoassay and the expression of steroidogenic genes by reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR). RESULTS: The sequencing of the RT-PCR amplicons confirmed the presence of both gonadotrophin receptors and immunohistochemistry localized them on the membrane of endometrial glands cells throughout the glandular epithelium. The gonadotrophin-receptor complex was able to increase the intracellular cAMP in a dose-response and time-course manner and to induce steroidogenic genes expression. CONCLUSION: This study demonstrates that both gonadotrophin receptors are expressed along the glandular epithelium of endometria and they mediate the effects of gonadotrophins on intracellular functions.


Subject(s)
Endometrium/metabolism , Receptors, FSH/genetics , Receptors, LH/genetics , Adult , Chorionic Gonadotropin/genetics , Cyclic AMP/metabolism , Endometrium/growth & development , Female , Follicle Stimulating Hormone/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Pregnancy , Receptors, FSH/metabolism , Signal Transduction/genetics
20.
Cell Physiol Biochem ; 43(3): 1064-1076, 2017.
Article in English | MEDLINE | ID: mdl-28977799

ABSTRACT

BACKGROUND/AIMS: Physiological role of luteinizing hormone (LH) and its receptor (LHCGR) in adrenal remains unknown. In inhibin-α/Simian Virus 40 T antigen (SV40Tag) (inhα/Tag) mice, gonadectomy-induced (OVX) elevated LH triggers the growth of transcription factor GATA4 (GATA4)-positive adrenocortical tumors in a hyperplasia-adenoma-adenocarcinoma sequence. METHODS: We investigated the role of LHCGR in tumor induction, by crossbreeding inhα/Tag with Lhcgr knockout (LuRKO) mice. By knocking out Lhcgr and Gata4 in Cα1 adrenocortical cells (Lhcgr-ko, Gata4-ko) we tested their role in tumor progression. RESULTS: Adrenal tumors of OVX inhα/Tag mice develop from the hyperplastic cells localized in the topmost layer of zona fasciculata. OVX inhα/Tag/LuRKO only developed SV40Tag positive hyperplastic cells that were GATA4 negative, cleaved caspase-3 positive and did not progress into adenoma. In contrast to Lhcgr-ko, Gata4-ko Cα1 cells presented decreased proliferation, increased apoptosis, decreased expression of Inha, SV40Tag and Lhcgr tumor markers, as well as up-regulated adrenal- and down-regulated sex steroid gene expression. Both Gata4-ko and Lhcgr-ko Cα1 cells had decreased expression of steroidogenic genes resulting in decreased basal progesterone production. CONCLUSION: Our data indicate that LH/LHCGR signaling is critical for the adrenal cell reprogramming by GATA4 induction prompting adenoma formation and gonadal-like phenotype of the adrenocortical tumors in inhα/Tag mice.


Subject(s)
Adrenal Cortex Neoplasms/pathology , GATA4 Transcription Factor/metabolism , Luteinizing Hormone/metabolism , Adrenal Cortex Neoplasms/etiology , Adrenal Cortex Neoplasms/metabolism , Adrenal Glands/metabolism , Adrenal Glands/pathology , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Apoptosis , CRISPR-Cas Systems/genetics , Caspase 3/metabolism , Cell Proliferation , Cell Transformation, Neoplastic , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Down-Regulation , Female , Fluoroimmunoassay , GATA4 Transcription Factor/deficiency , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gonads/surgery , Inhibins/genetics , Inhibins/metabolism , Luteinizing Hormone/blood , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , Phosphoproteins/metabolism , Receptors, LH/deficiency , Receptors, LH/genetics , Steroidogenic Factor 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL