Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.274
Filter
Add more filters

Publication year range
1.
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
2.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Article in English | MEDLINE | ID: mdl-33500349

ABSTRACT

The gonadotropin-releasing hormone (GnRH) pulse is fundamental for mammalian reproduction: GnRH pulse regimens are needed as therapies for infertile women as continuous GnRH treatment paradoxically inhibits gonadotropin release. Circumstantial evidence suggests that the hypothalamic arcuate KNDy neurons expressing kisspeptin (encoded by Kiss1), neurokinin B (encoded by Tac3), and dynorphin A serve as a GnRH pulse generator; however, no direct evidence is currently available. Here, we show that rescuing >20% KNDy neurons by transfecting Kiss1 inside arcuate Tac3 neurons, but not outside of these neurons, recovered folliculogenesis and luteinizing hormone (LH) pulses, an indicator of GnRH pulses, in female global Kiss1 knockout (KO) rats and that >90% conditional arcuate Kiss1 KO in newly generated Kiss1-floxed rats completely suppressed LH pulses. These results first provide direct evidence that KNDy neurons are the GnRH pulse generator, and at least 20% of KNDy neurons are sufficient to maintain folliculogenesis via generating GnRH/gonadotropin pulses.


Subject(s)
Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropins/metabolism , Kisspeptins/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Organogenesis , Ovarian Follicle/growth & development , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Aromatase/genetics , Aromatase/metabolism , Feedback, Physiological , Female , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Integrases/metabolism , Luteinizing Hormone/blood , Organ Size , Ovarian Follicle/metabolism , Pituitary Gland/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, LH/genetics , Receptors, LH/metabolism , Receptors, LHRH/metabolism
3.
Zhongguo Zhong Yao Za Zhi ; 49(12): 3288-3294, 2024 Jun.
Article in Zh | MEDLINE | ID: mdl-39041091

ABSTRACT

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


Subject(s)
Drugs, Chinese Herbal , Dysmenorrhea , Ovary , Animals , Female , Mice , Ovary/drug effects , Ovary/metabolism , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/pharmacology , Dysmenorrhea/drug therapy , Dysmenorrhea/metabolism , Dysmenorrhea/genetics , Dysmenorrhea/physiopathology , Luteinizing Hormone/blood , Follicle Stimulating Hormone/blood , Pituitary Gland/metabolism , Pituitary Gland/drug effects , Humans , Receptors, FSH/genetics , Receptors, FSH/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism
4.
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
5.
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
6.
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
7.
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
8.
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
9.
EMBO J ; 37(17)2018 09 03.
Article in English | MEDLINE | ID: mdl-30037826

ABSTRACT

The number and self-renewal capacity of hematopoietic stem cells (HSCs) are tightly regulated at different developmental stages. Many pathways have been implicated in regulating HSC development in cell autonomous manners; however, it remains unclear how HSCs sense and integrate developmental cues. In this study, we identified an extrinsic mechanism by which HSC number and functions are regulated during mouse puberty. We found that the HSC number in postnatal bone marrow reached homeostasis at 4 weeks after birth. Luteinizing hormone, but not downstream sex hormones, was involved in regulating HSC homeostasis during this period. Expression of luteinizing hormone receptor (Lhcgr) is highly restricted in HSCs and multipotent progenitor cells in the hematopoietic hierarchy. When Lhcgr was deleted, HSCs continued to expand even after 4 weeks after birth, leading to abnormally elevated hematopoiesis and leukocytosis. In a murine acute myeloid leukemia model, leukemia development was significantly accelerated upon Lhcgr deletion. Together, our work reveals an extrinsic counting mechanism that restricts HSC expansion during development and is physiologically important for maintaining normal hematopoiesis and inhibiting leukemogenesis.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/metabolism , Luteinizing Hormone/metabolism , Receptors, LH/metabolism , Sexual Maturation , Signal Transduction , Animals , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Luteinizing Hormone/genetics , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Receptors, LH/genetics
10.
Reprod Biol Endocrinol ; 20(1): 43, 2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35236366

ABSTRACT

The heavy metal cadmium is proposed to be one of the environmental endocrine disruptors of spermatogenesis. Cadmium-induced inhibition of spermatogenesis is associated with a hormone secretion disorder. Letrozole is an aromatase inhibitor that increases peripheral androgen levels and stimulates spermatogenesis. However, the potential protective effects of letrozole on cadmium-induced reproductive toxicity remain to be elucidated. In this study, male mice were administered CdCl2 (4 mg/kg BW) orally by gavage alone or in combination with letrozole (0.25 mg/kg BW) for 30 days. Cd exposure caused a significant decreases in body weight, sperm count, motility, vitality, and plasma testosterone levels. Histopathological changes revealed extensive vacuolization and decreased spermatozoa in the lumen. However, in the Cd + letrozole group, letrozole treatment compensated for deficits in sperm parameters (count, motility, and vitality) induced by Cd. Letrozole treatment significantly increased serum testosterone levels, which were reduced by Cd. Histopathological studies revealed a systematic array of all germ cells, a preserved basement membrane and relatively less vacuolization. For a mechanistic examination, RNA-seq was used to profile alterations in gene expression in response to letrozole. Compared with that in the Cd-treated group, RNA-Seq analysis showed that 214 genes were differentially expressed in the presence of letrozole. Gene ontology (GO) enrichment analysis and KEGG signaling pathway analysis showed that steroid biosynthetic processes were the processes most affected by letrozole treatment. Furthermore, we found that the expression of the testosterone synthesis-related genes LHCGR (luteinizing hormone/choriogonadotropin receptor) and Hsd3b6 (3 beta- and steroid delta-isomerase 6) was significantly downregulated in Cd-treated testes, but these genes maintained similar expression levels in letrozole-treated testes as those in the control group. However, the transcription levels of inflammatory cytokines, such as IL-1ß and IL-6, and oxidative stress-related genes (Nrf2, Nqo1, and Ho-1) showed no changes. The present study suggests that the potential protective effect of letrozole on Cd-induced reproductive toxicity might be mediated by the upregulation of LHCGR and Hsd3b6, which would beneficially increase testosterone synthesis to achieve optimum protection of sperm quality and spermatogenesis.


Subject(s)
Cadmium , Letrozole , Spermatogenesis , Testosterone , Animals , Male , Mice , Cadmium/toxicity , Cytoprotection/drug effects , Cytoprotection/genetics , Letrozole/pharmacology , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mice, Inbred ICR , Protective Agents/pharmacology , Receptors, LH/drug effects , Receptors, LH/genetics , Receptors, LH/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Spermatogenesis/drug effects , Spermatogenesis/genetics , Spermatozoa/drug effects , Spermatozoa/metabolism , Steroid Isomerases/drug effects , Steroid Isomerases/genetics , Steroid Isomerases/metabolism , Testis/drug effects , Testis/metabolism , Testosterone/biosynthesis
11.
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
12.
Zygote ; 30(4): 577-583, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35499099

ABSTRACT

The development of an ovarian follicle is a complex process at the cellular and molecular level that is mainly regulated by follicle-stimulating hormone receptor (FSHR) and luteinizing hormone receptor (LHR). To elucidate the contribution of these receptors to ovarian follicle development, it is necessary to determine their expression profiles during this biological process. Therefore, this study aimed to investigate the relationship between ovarian development pattern and the differential ovarian expression pattern of FSHR and LHR genes as well as proteins at different developmental stages. Ovaries were collected from 30 New Zealand rabbits at day 0 (birth), week 2 (neonate), week 4 (cub), week 16 (maturity), and day 18 pregnancy. Ovarian histology, and gene as well as protein expression were determined using light microscopy, real-time PCR and western blotting, respectively. The results showed that the expression levels of FSHR mRNA and protein increased coincidently with age and the growth of ovarian follicles. The levels of LHR mRNA and protein remained low from the day of birth until week 4 and became significantly higher by week 16 coinciding with appearance of growing and antral follicles, which have a defined thecal layer. FSHR gene and protein expression decreased with pregnancy, whereas LHR increased, reaching a peak level during pregnancy. It can be concluded that changes in FSHR and LHR gene and protein expression could be related to the growth and development of follicles, indicating the regulatory role for these receptors in rabbit folliculogenesis.


Subject(s)
Receptors, FSH , Receptors, LH , Animals , Female , Ovarian Follicle , Ovary/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Receptors, FSH/genetics , Receptors, LH/genetics
13.
Int J Mol Sci ; 23(24)2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36555365

ABSTRACT

In the mouse, two distinct populations of Leydig cells arise during testis development. Fetal Leydig cells arise from a stem cell population and produce T required for masculinization. It is debated whether they persist in the adult testis. A second adult Leydig stem cell population gives rise to progenitor-immature-mature adult type Leydig cells that produce T in response to LH to maintain spermatogenesis. In testis of adult null male mice lacking either only LH (Lhb-/-) or LHR (Lhr-/-), mature Leydig cells are absent but fetal Leydig cells persist. Thus, it is not clear whether other ligands signal via LHRs in Lhb null mice or LH signals via other receptors in the absence of LHR in Lhr null mice. Moreover, it is not clear whether truncated LHR isoforms generated from the same Lhr gene promoter encode functionally relevant LH receptors. To determine the in vivo roles of LH-LHR signaling pathway in the Leydig cell lineage, we generated double null mutant mice lacking both LH Ligand and all forms of LHR. Phenotypic analysis indicated testis morpho-histological characteristics are identical among double null and single mutants which all showed poorly developed interstitium with a reduction in Leydig cell number and absence of late stage spermatids. Gene expression analyses confirmed that the majority of the T biosynthesis pathway enzyme-encoding mRNAs expressed in Leydig cells were all suppressed. Expression of thrombospondin-2, a fetal Leydig cell marker gene was upregulated in single and double null mutants indicating that fetal Leydig cells originate and develop independent of LH-LHR signaling pathway in vivo. Serum and intratesticular T levels were similarly suppressed in single and double mutants. Consequently, expression of AR-regulated genes in Sertoli and germ cells were similarly affected in single and double mutants without any evidence of any additive effect in the combined absence of both LH and LHR. Our studies unequivocally provide genetic evidence that in the mouse testis, fetal Leydig cells do not require LH-LHR signaling pathway and a one-to-one LH ligand-LHR signaling pathway exists in vivo to regulate adult Leydig cell lineage and spermatogenesis.


Subject(s)
Leydig Cells , Testis , Mice , Male , Animals , Leydig Cells/metabolism , Ligands , Testis/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Signal Transduction , Testosterone/metabolism
14.
Int J Mol Sci ; 23(20)2022 Oct 18.
Article in English | MEDLINE | ID: mdl-36293309

ABSTRACT

Under stress conditions, luteinizing hormone (LH)-mediated ovulation is inhibited, resulting in insufficient oocyte production and excretion during follicular development. When the body is stressed, a large amount of corticosterone (CORT) is generated, which will lead to a disorder of the body's endocrine system and damage to the body. Our previous work showed that CORT can block follicular development in mice. Since LH acts through binding with the luteinizing hormone receptor (Lhcgr), the present study aimed to investigate whether and how corticosterone (CORT) influences Lhcgr expression in mouse ovarian granulosa cells (GCs). For this purpose, three-week-old ICR female mice were injected intraperitoneally with pregnant mare serum gonadotropin (PMSG). In addition, the treatment group was injected with CORT (1 mg/mouse) at intervals of 8 h and the control group was injected with the same volume of methyl sulfoxide (DMSO). GCs were collected at 24 h, 48 h, and 55 h after PMSG injection. For in vitro experiments, the mouse GCs obtained from healthy follicles were treated with CORT alone, or together with inhibitors against the glucocorticoid receptor (Nr3c1). The results showed that the CORT caused a downregulation of Lhcgr expression in GCs, which was accompanied by impaired cell viability. Moreover, the effect of the CORT was mediated by binding to its receptor (Nr3c1) in GCs. Further investigation revealed that Nr3c1 might regulate the transcription of Lhcgr through inhibiting the expression of Lhcgr transcription factors, including AP1 and Creb. Taken together, our findings suggested a possible mechanism of CORT-induced anovulation involving the inhibition of Lhcgr expression in GCs by the CORT-Nr3c1-AP1/Creb axis.


Subject(s)
Corticosterone , Receptors, LH , Horses , Female , Mice , Animals , Receptors, LH/genetics , Receptors, LH/metabolism , Corticosterone/pharmacology , Corticosterone/metabolism , Gonadotropins, Equine/metabolism , Gonadotropins, Equine/pharmacology , Receptors, Glucocorticoid/metabolism , Granulosa Cells/metabolism , Glucocorticoids/metabolism , Dimethyl Sulfoxide/pharmacology , Mice, Inbred ICR , Luteinizing Hormone/pharmacology , Luteinizing Hormone/metabolism , Receptors, G-Protein-Coupled/metabolism , Transcription Factors/metabolism
15.
Bull Exp Biol Med ; 172(4): 435-440, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35175480

ABSTRACT

We compared the effectiveness of human chorionic gonadotropin (hCG; 5 days, 20 IU/rat/day), allosteric luteinizing hormone receptor agonist TP04 (5 days, 20 mg/kg/day), and metformin (28 days, 120 mg/kg/day) in restoring spermatogenesis in male rats with type 2 diabetes mellitus. hCG and TP04 increased the levels of testosterone and expression of the steroidogenic protein StAR, the number of spermatogenic cells, thickness of the seminal epithelium, and the number and motility of mature sperm that were reduced in diabetic rats, though they did not reduce the number of defective spermatozoa. Metformin had a weak effect on steroidogenesis, but was not inferior to luteinizing hormone receptor agonist by its restorative effect on spermatogenesis and also reduced the number of defective forms of spermatozoa. Thus, the spermatogenesis-restoring effect of metformin and luteinizing hormone receptor agonist in type 2 diabetes mellitus are comparable, despite different mechanisms of action.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Metformin , Animals , Chorionic Gonadotropin/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Male , Metformin/pharmacology , Rats , Receptors, LH/agonists , Receptors, LH/genetics , Receptors, LH/metabolism , Spermatogenesis , Streptozocin , Testis/metabolism , Testosterone/metabolism
16.
BMC Biotechnol ; 21(1): 52, 2021 09 05.
Article in English | MEDLINE | ID: mdl-34482828

ABSTRACT

BACKGROUND: Equine chorionic gonadotropin (eCG), which comprises highly glycosylated α-subunit and ß-subunit, is a unique member of the glycoprotein hormone family as it elicits both follicle-stimulating hormone (FSH)-like and luteinizing hormone (LH)-like responses in non-equid species. To examine the biological function of glycosylated sites in eCG, the following glycosylation site mutants were constructed: eCGß/αΔ56, substitution of Asn56 of α-subunit with Gln; eCGß-D/α, deletion of the O-linked glycosylation site at the carboxyl-terminal peptide (CTP) region of the ß-subunit; eCGß-D/αΔ56, double mutant. The recombinant eCG (rec-eCG) mutants were expressed in Chinese hamster ovary suspension (CHO-S) cells. The FSH-like and LH-like activities of the mutants were examined using CHO-K1 cells expressing rat lutropin/CG receptor (rLH/CGR) and rat FSH receptor (rFSHR). RESULTS: Both rec-eCGß/α and rec-eCGß/αΔ56 were efficiently secreted into the CHO-S cell culture medium on day 1 post-transfection. However, the secretion of eCGß-D/α and eCGß-D/αΔ56, which lack approximately 12 O-linked glycosylation sites, was slightly delayed. The expression levels of all mutants were similar (200-250 mIU/mL) from days 3 to 7 post-transfection. The molecular weight of rec-eCGß/α, rec-eCGß/αΔ56 and rec-eCG ß-D/α were in the ranges of 40-45, 37-42, and 34-36 kDa, respectively. Treatment with peptide-N-glycanase F markedly decreased the molecular weight to approximately 5-10 kDa. Rec-eCGß/αΔ56 exhibited markedly downregulated LH-like activity. The signal transduction activity of both double mutants was completely impaired. This indicated that the glycosylation site at Asn56 of the α-subunit plays a pivotal role in the LH-like activity of eCG. Similarly, the FSH-like activity of the mutants was markedly downregulated. eCGß-D/α exhibited markedly downregulated LH-like and FSH-like activities. CONCLUSIONS: Rec-eCGß/α exhibits potent biological activity in cells expressing rLH/CGR and rFSHR. The findings of this study suggest that the LH-like and FSH-like activities of eCG are regulated by the N-linked glycosylation site at Asn56 of the eCG α-subunit and/or by the O-linked glycosylation sites of the eCG ß-subunit. These findings improved our understanding of the mechanisms underlying both LH-like and FSH-like activities of eCG.


Subject(s)
Chorionic Gonadotropin , Luteinizing Hormone , Animals , CHO Cells , Chorionic Gonadotropin/metabolism , Cricetinae , Cricetulus , Glycosylation , Horses , Luteinizing Hormone/metabolism , Rats , Receptors, FSH/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Signal Transduction
17.
Mol Hum Reprod ; 27(6)2021 05 29.
Article in English | MEDLINE | ID: mdl-33905521

ABSTRACT

Secreted frizzled-related protein-4 (SFRP4) belongs to a family of soluble ovarian-expressed proteins that participate in female reproduction, particularly in rodents. In humans, SFRP4 is highly expressed in cumulus cells (CCs). However, the mechanisms that stimulate SFRP4 in CCs have not been examined. We hypothesise that oocyte-secreted factors such as growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) are involved in the regulation of SFRP4. Human CCs were collected from patients undergoing fertility treatments and treated with GDF9 or BMP15 or their combination in the presence of FSH or vehicle. FSH treatment significantly decreased SFRP4 mRNA levels when compared with nontreated cells. However, SFRP4 mRNA levels were increased significantly by GDF9 plus BMP15 in a concentration-dependent manner in the presence or absence of FSH. The combination of GDF9 plus BMP15 also increased SFRP4 protein levels and decreased the activity of the ß-catenin/T cell factor-responsive promoter significantly. GDF9 plus BMP15 inhibited steroidogenic acute regulatory protein and LH/hCG receptor stimulation by FSH, while treatment with SFRP4 blocked the stimulatory effect of FSH on these genes. The evidence demonstrates that GDF9 and BMP15 act in coordination to stimulate SFRP4 expression and suggests that SFRP4 mediates the anti-luteinising effects of the oocyte in human CCs.


Subject(s)
Bone Morphogenetic Protein 15/pharmacology , Cumulus Cells/drug effects , Growth Differentiation Factor 9/pharmacology , Intercellular Signaling Peptides and Proteins/physiology , Oocytes/physiology , Proto-Oncogene Proteins/biosynthesis , Bone Morphogenetic Protein 15/administration & dosage , Cells, Cultured , Cumulus Cells/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Female , Follicle Stimulating Hormone/pharmacology , Gene Expression Regulation/drug effects , Genes, Reporter , Growth Differentiation Factor 9/administration & dosage , Humans , Oocytes/chemistry , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Primary Cell Culture , Proto-Oncogene Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, LH/biosynthesis , Receptors, LH/genetics , Species Specificity
18.
Cell Commun Signal ; 19(1): 8, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33478524

ABSTRACT

BACKGROUND: First identified as a regulator of neuronal axon guidance, Slit/Robo signaling has since been implicated in additional physiologic and pathologic processes, such as angiogenesis, organogenesis and cancer progression. However, its roles in the regulation of testis function have been little explored. METHODS: Immunohistochemistry and RT-qPCR analyses were performed to detect the expression of Slit/Robo signaling effectors in the adult mouse testis. To identify the roles and mechanisms of Slit/Robo signaling in the regulation of steroidogenesis, RT-qPCR, immunoblotting and hormone measurements were carried out using Leydig cells (primary cultures and the MA10 cell line) treated with exogenous SLIT ligands, and testes from Robo1-null mice. RESULTS: Slit1, -2 and -3 and Robo1 and -2 expression was detected in the adult mouse testis, particularly in Leydig cells. In vitro treatment of Leydig cells with exogenous SLIT ligands led to a decrease in the expression of the steroidogenic genes Star, Cyp11a1, and Cyp17a1. SLIT2 treatment decreased the phosphorylation of the key steroidogenic gene regulator CREB, possibly in part by suppressing AKT activity. Furthermore, SLIT2 treatment reduced the responsiveness of MA10 cells to luteinizing hormone by decreasing the expression of Lhcgr. Consistent with these in vitro results, an increase in testicular Star mRNA levels and intra-testicular testosterone concentrations were found in Robo1-null mice. Finally, we showed that the expression of the Slit and Robo genes in Leydig cells is enhanced by testosterone treatment in vitro, by an AR-independent mechanism. CONCLUSION: Taken together, these results suggest that Slit/Robo signaling represents a novel mechanism that regulates Leydig cell steroidogenesis. It may act in an autocrine/paracrine manner to mediate negative feedback by testosterone on its own synthesis. Video Abstract.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Leydig Cells/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Testosterone/biosynthesis , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Luteinizing Hormone/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Progesterone/biosynthesis , Receptors, Immunologic/genetics , Receptors, LH/genetics , Signal Transduction
19.
Horm Behav ; 127: 104877, 2021 01.
Article in English | MEDLINE | ID: mdl-33186586

ABSTRACT

The evolution of social behavior depends on genetic changes, yet, how genomic variation manifests itself in behavioral diversity is still largely unresolved. Chromosomal inversions can play a pivotal role in producing distinct behavioral phenotypes, in particular, when inversion genes are functionally associated with hormone synthesis and signaling. Male ruffs exhibit alternative reproductive tactics (ARTs) with an autosomal inversion determining two alternative morphs with clear behavioral and hormonal differences from the ancestral morph. We investigated hormonal and transcriptomic differences in the pituitary and gonads. Using a GnRH challenge, we found that the ability to synthesize testosterone in inversion carriers is severely constrained, whereas the synthesis of androstenedione, a testosterone precursor, is not. Inversion morphs were able to produce a transient increase in androstenedione following the GnRH injection, supporting the view that pituitary sensitivity to GnRH is comparable to that of the ancestral morph. We then performed gene expression analyses in a second set of untreated birds and found no evidence of alterations to pituitary sensitivity, gonadotropin production or gonad sensitivity to luteinizing hormone or follicle-stimulating hormone across morphs. Inversion morphs also showed reduced progesterone receptor expression in the pituitary. Strikingly, in the gonads, inversion morphs over-expressed STAR, a gene that is located outside of the inversion and responsible for providing the cholesterol substrate required for the synthesis of sex hormones. In conclusion, our results suggest that the gonads determine morph-specific differences in hormonal regulation.


Subject(s)
Charadriiformes/physiology , Polymorphism, Genetic , Reproduction/genetics , Androstenedione/metabolism , Animals , Charadriiformes/genetics , Follicle Stimulating Hormone, beta Subunit/genetics , Follicle Stimulating Hormone, beta Subunit/metabolism , Gene Expression/drug effects , Gonadal Steroid Hormones/biosynthesis , Gonadotropin-Releasing Hormone/pharmacology , Gonads/drug effects , Gonads/metabolism , Gonads/physiology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiology , Male , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Receptors, LH/genetics , Receptors, LH/metabolism , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Reproduction/drug effects , Sequence Inversion , Sexual Behavior, Animal/drug effects , Sexual Behavior, Animal/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Testosterone/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL