Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.270
Filtrar
1.
Endocrinology ; 165(6)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38679471

RESUMEN

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.


Asunto(s)
Proteínas Fluorescentes Verdes , Unión Proteica , Humanos , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Fluorescentes Verdes/genética , Receptores de HL/metabolismo , Receptores de HL/genética , Luciferasas/metabolismo , Luciferasas/genética , Animales , Transferencia de Energía por Resonancia de Bioluminiscencia/métodos , Gonadotropina Coriónica/metabolismo , Células HEK293 , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/genética , Transferencia de Energía , Glicoproteínas/metabolismo , Mediciones Luminiscentes/métodos
2.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167165, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38653355

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer , Gonadotropina Coriónica , Modelos Animales de Enfermedad , Receptores de HL , Animales , Femenino , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Ratones , Gonadotropina Coriónica/farmacología , Receptores de HL/metabolismo , Receptores de HL/genética , Receptores de HL/agonistas , Ratones Transgénicos , Ovariectomía , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Humanos , Reproducción/efectos de los fármacos , Presenilina-1/genética , Presenilina-1/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Cognición/efectos de los fármacos
3.
Endocrine ; 83(2): 473-482, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37828397

RESUMEN

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.


Asunto(s)
Síndrome de Cushing , Femenino , Embarazo , Humanos , Síndrome de Cushing/diagnóstico , Receptores de HL/genética , Receptores de HL/metabolismo , Hormona Luteinizante/metabolismo , Gonadotropina Coriónica , Mutación , Hidrocortisona , Cromograninas/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética
4.
J Pak Med Assoc ; 73(12): 2458-2461, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38083932

RESUMEN

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.


Asunto(s)
Pubertad Precoz , Niño , Humanos , Lactante , Masculino , Gonadotropina Coriónica , Mutación , Mutación Missense , Pakistán , Pubertad Precoz/genética , Receptores de HL/genética
5.
Adv Sci (Weinh) ; 10(29): e2300993, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37697644

RESUMEN

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.


Asunto(s)
Hipogonadismo , Células Intersticiales del Testículo , Receptores de HL , Animales , Humanos , Masculino , Ratones , Diferenciación Celular , Hipogonadismo/genética , Hipogonadismo/terapia , Células Intersticiales del Testículo/trasplante , Mutación , Receptores Acoplados a Proteínas G , Testosterona/metabolismo , Receptores de HL/genética
6.
J Ovarian Res ; 16(1): 189, 2023 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-37691102

RESUMEN

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.


Asunto(s)
Hormona Luteinizante , Oocitos , Receptores de HL , Humanos , Gonadotropina Coriónica , Hormona Liberadora de Gonadotropina/genética , Receptores Acoplados a Proteínas G , Estudios Retrospectivos , Receptores de HL/genética
7.
Int J Mol Sci ; 24(15)2023 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-37569429

RESUMEN

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.


Asunto(s)
Kisspeptinas , Receptores de HL , Ratones , Animales , Humanos , Receptores de HL/genética , Receptores de HL/metabolismo , Kisspeptinas/metabolismo , Ligandos , AMP Cíclico/metabolismo , Transducción de Señal , Receptores Acoplados a Proteínas G , Hormona Folículo Estimulante/farmacología , Hormona Folículo Estimulante/metabolismo , Tirotropina/metabolismo , Gonadotropina Coriónica/metabolismo
8.
Res Vet Sci ; 161: 132-137, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37384971

RESUMEN

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.


Asunto(s)
Células Intersticiales del Testículo , Receptores de HL , Masculino , Animales , Receptores de HL/genética , Receptores de HL/metabolismo , Testosterona/metabolismo , Testículo/metabolismo , Hormona Luteinizante/farmacología , Hormona Luteinizante/metabolismo , Mamíferos
9.
Biochem Genet ; 61(4): 1418-1432, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36633772

RESUMEN

 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.


Asunto(s)
Síndrome del Ovario Poliquístico , Receptores de HL , Humanos , Femenino , Adulto , Receptores de HL/genética , Síndrome del Ovario Poliquístico/genética , Predisposición Genética a la Enfermedad , Estudios de Casos y Controles , Frecuencia de los Genes , Hormona Luteinizante/genética , Polimorfismo de Nucleótido Simple
10.
J Endocrinol ; 257(1)2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36705256

RESUMEN

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.


Asunto(s)
Etanol , Hormona Folículo Estimulante , Ratas , Femenino , Animales , Etanol/farmacología , Etanol/metabolismo , Hormona Folículo Estimulante/farmacología , Hormona Folículo Estimulante/metabolismo , Células de la Granulosa/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , ARN Mensajero/metabolismo , Células Cultivadas , Estrógenos/farmacología , Estrógenos/metabolismo
11.
Biol Reprod ; 108(2): 258-268, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36214501

RESUMEN

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.


Asunto(s)
Células Endoteliales , Neovascularización Fisiológica , Ovario , Receptores de HL , Animales , Femenino , Humanos , Gonadotropina Coriónica/farmacología , Gonadotropina Coriónica/metabolismo , Células Endoteliales/metabolismo , Hormona Luteinizante/farmacología , Hormona Luteinizante/metabolismo , Macaca fascicularis , Neovascularización Fisiológica/fisiología , Folículo Ovárico/metabolismo , Ovario/irrigación sanguínea , Ovario/metabolismo , Ovulación/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo
12.
Anim Biotechnol ; 34(7): 2285-2294, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35714982

RESUMEN

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.


Asunto(s)
Kisspeptinas , Vacunas de ADN , Ovinos/genética , Animales , Masculino , Kisspeptinas/genética , Receptores de Kisspeptina-1 , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Hormona Liberadora de Gonadotropina/genética , Oveja Doméstica/genética , Receptores Acoplados a Proteínas G/genética , Receptores de HL/genética , Inmunización/veterinaria , ARN Mensajero/genética , ARN Mensajero/metabolismo , Hormona Folículo Estimulante/genética
13.
Int J Mol Sci ; 23(24)2022 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-36555365

RESUMEN

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.


Asunto(s)
Células Intersticiales del Testículo , Testículo , Ratones , Masculino , Animales , Células Intersticiales del Testículo/metabolismo , Ligandos , Testículo/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Transducción de Señal , Testosterona/metabolismo
14.
BMC Endocr Disord ; 22(1): 335, 2022 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-36585675

RESUMEN

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.


Asunto(s)
Síndrome del Ovario Poliquístico , Receptores de HL , Femenino , Humanos , Estudios de Casos y Controles , Colesterol , Predisposición Genética a la Enfermedad , Lipoproteínas LDL , Hormona Luteinizante , Síndrome del Ovario Poliquístico/genética , Polimorfismo de Nucleótido Simple , Receptores Acoplados a Proteínas G/genética , Receptores de HL/genética , Testosterona , Triglicéridos
15.
Am J Vet Res ; 83(12)2022 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-36327166

RESUMEN

OBJECTIVE: To determine luteinizing hormone receptor (LHR) expression and response to LHR activation in isolated canine splenic hemangiosarcoma cell lines in vitro. SAMPLES: In vitro cultures of commercially available canine splenic hemangiosarcoma cell lines (EFS, GRACE-HSA, and DAL-4). PROCEDURES: The percentage of each cell line expressing LHR was determined by immunocytochemistry. Cells were then treated with increasing doses (7.5 ng/mL, 75 ng/mL) of recombinant canine luteinizing hormone (cLH) for 48 hours and evaluated using a cell proliferation assay. RESULTS: The percentage of cells expressing LHR was 17.2 ± 4.5%, 11.8 ± 3.1%, and 6.9 ± 2.5% in EFS, GRACE-HSA, and DAL-4, respectively. There was significant increase in cell count in the DAL-4 and EFS cell lines following a 48-hour incubation at the highest cLH concentration (P = .028 and P = .019, respectively). There was not a significant increase in cell count in the GRACE-HSA cell line at either cLH concentration. CLINICAL RELEVANCE: Activation of LHR results in cell proliferation in some canine splenic hemangiosarcoma cell lines. These results may explain why spayed and castrated dogs with high circulating LH concentrations may develop hemangiosarcoma more frequently than intact dogs.


Asunto(s)
Enfermedades de los Perros , Hemangiosarcoma , Neoplasias del Bazo , Perros , Animales , Hemangiosarcoma/veterinaria , Hemangiosarcoma/tratamiento farmacológico , Receptores de HL/genética , Enfermedades de los Perros/tratamiento farmacológico , Neoplasias del Bazo/veterinaria , Línea Celular Tumoral
16.
Braz J Biol ; 82: e262017, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36197367

RESUMEN

The aim of the current study is to evaluate gene expression patterns of LH (lhr) and estrogen (er) receptors and plasma steroid levels during testicular development in Genyatremus luteus. Males were histologically classified as immature (n=7), maturing (n=7) and mature (n=7), based on the cellular structure of their testes. Plasma 11-KT concentration recorded peak at the final maturation stage. The highest plasma 17α-OHP concentrations were observed at the immature stage; they decreased at the maturation and mature stages. On the other hand, 17ß-estradiol (E2) recorded higher concentrations at the maturation stage. Er expression has significantly increased along the maturational development of animals' testes. The mRNA observed for the LH receptor has decreased from immature to maturing stage; it presented expression peak at the mature stage. There was high association between receptor gene expression and plasma steroid levels, mainly E2. The current study was the first to feature different reproductive maturation stages in male G. luteus specimens, based on cellular, endocrine and molecular aspects. In addition, it has shown that the gene expression profile for er and lhr receptors, as well as plasma 11-KT and E2 concentrations, are directly linked to testicular maturation, although they are not necessarily associated with the gonadosomatic index.


Asunto(s)
Perciformes , Receptores de HL , Animales , Estradiol , Estrógenos , Peces , Expresión Génica , Hormonas Esteroides Gonadales , Masculino , Perciformes/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de HL/genética
17.
Int J Mol Sci ; 23(20)2022 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-36293309

RESUMEN

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.


Asunto(s)
Corticosterona , Receptores de HL , Caballos , Femenino , Ratones , Animales , Receptores de HL/genética , Receptores de HL/metabolismo , Corticosterona/farmacología , Corticosterona/metabolismo , Gonadotropinas Equinas/metabolismo , Gonadotropinas Equinas/farmacología , Receptores de Glucocorticoides/metabolismo , Células de la Granulosa/metabolismo , Glucocorticoides/metabolismo , Dimetilsulfóxido/farmacología , Ratones Endogámicos ICR , Hormona Luteinizante/farmacología , Hormona Luteinizante/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Factores de Transcripción/metabolismo
18.
Cell Rep Med ; 3(11): 100792, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36270285

RESUMEN

Leydig cell failure (LCF) caused by gene mutation results in testosterone deficiency and infertility. Serum testosterone levels can be recovered via testosterone replacement; however, established therapies have shown limited success in restoring fertility. Here, we use a luteinizing hormone/choriogonadotrophin receptor (Lhcgr)-deficient mouse model of LCF to investigate the feasibility of gene therapy for restoring testosterone production and fertility. We screen several adeno-associated virus (AAV) serotypes and identify AAV8 as an efficient vector to drive exogenous Lhcgr expression in progenitor Leydig cells through interstitial injection. We observe considerable testosterone recovery and Leydig cell maturation after AAV8-Lhcgr treatment in pubertal Lhcgr-/- mice. Of note, this gene therapy partially recovers sexual development, substantially restores spermatogenesis, and effectively produces fertile offspring. Furthermore, these favorable effects can be reproduced in adult Lhcgr-/- mice. Our proof-of-concept experiments in the mouse model demonstrate that AAV-mediated gene therapy may represent a promising therapeutic approach for patients with LCF.


Asunto(s)
Células Intersticiales del Testículo , Receptores de HL , Masculino , Ratones , Animales , Células Intersticiales del Testículo/metabolismo , Receptores de HL/genética , Dependovirus/genética , Gonadotropina Coriónica/genética , Testosterona , Fertilidad/genética , Modelos Animales de Enfermedad , Terapia Genética
19.
Chemosphere ; 308(Pt 3): 136424, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36116629

RESUMEN

Bisphenol AF (BPAF) is one of the substitutes for bisphenol A (BPA), which has endocrine-disrupting, reproductive and neurological toxicity. BPAF has frequently been detected in the aquatic environment, which has been a long-term threat to the health of aquatic organisms. In this study, female marine medaka (Oryzias melastigma) were exposed to 6.7 µg/L, 73.4 µg/L, and 367.0 µg/L BPAF for 120 d. The effects of BPAF on behavior, growth, liver and ovarian histology, gene transcriptional profiles, and reproduction of marine medaka were determined. The results showed that with the increase of BPAF concentration, the swimming speed of female marine medaka showed an increasing trend and then decreasing trend. BPAF (367.0 µg/L) significantly increased body weight and condition factors in females. BPAF (73.4 µg/L and 367.0 µg/L) significantly delayed oocyte maturation. Exposure to 367.0 µg/L BPAF showed an increasing trend in the transcript levels of lipid synthesis and transport-related genes such as fatty acid synthase (fasn), sterol regulatory element binding protein (srebf), diacylglycerol acyltransferase (dgat), solute carrier family 27 member 4 (slc27a4), fatty acid-binding protein (fabp), and peroxisome proliferator-activated receptor gamma (pparγ) in the liver. In addition, 6.7 µg/L BPAF significantly down-regulated the expression levels of antioxidant-related genes [superoxide dismutase (sod), glutathione peroxidase (gpx), and catalase (cat)], and complement system-related genes [complement component 5 (c5), complement component 7a (c7a), mannan-binding lectin serine peptidase 1 (masp1), and tumor necrosis factor (tnf)] were significantly up-regulated in the 73.4 and 367.0 µg/L groups, which implies the effect of BPAF on the immune system in the liver. In the hypothalamic-pituitary-ovarian axis (HPG) results, the transcription levels of estrogen receptor α (erα), estrogen receptor ß (erß), androgen receptor (arα), gonadotropin-releasing hormone 2 (gnrh2), cytochrome P450 19b (cyp19b), aromatase (cyp19a), and luteinizing hormone receptor (lhr) in the brain and ovary, and vitellogenin (vtg) and choriogenin (chg) in the liver of 367.0 µg/L BPAF group showed a downward trend. In addition, exposure to 367.0 µg/L BPAF for 120 d inhibited the spawning behavior of marine medaka. Our results showed that long-term BPAF treatment influenced growth (body weight and condition factors), lipid metabolism, and ovarian maturation, and significantly altered the immune response and the transcriptional expression levels of HPG axis-related genes.


Asunto(s)
Lectina de Unión a Manosa , Oryzias , Contaminantes Químicos del Agua , Animales , Antioxidantes/metabolismo , Aromatasa/metabolismo , Compuestos de Bencidrilo , Peso Corporal , Catalasa/metabolismo , Complemento C5/genética , Complemento C5/metabolismo , Diacilglicerol O-Acetiltransferasa/genética , Diacilglicerol O-Acetiltransferasa/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Ácido Graso Sintasas/genética , Ácido Graso Sintasas/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Femenino , Fluorocarburos , Expresión Génica , Glutatión Peroxidasa/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Lípidos , Lectina de Unión a Manosa/genética , Lectina de Unión a Manosa/metabolismo , Oryzias/fisiología , PPAR gamma/metabolismo , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Receptores Androgénicos/metabolismo , Receptores de HL/genética , Serina/genética , Serina/metabolismo , Proteínas de Unión a los Elementos Reguladores de Esteroles/genética , Proteínas de Unión a los Elementos Reguladores de Esteroles/metabolismo , Superóxido Dismutasa/metabolismo , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/metabolismo , Vitelogeninas/metabolismo , Contaminantes Químicos del Agua/metabolismo , Contaminantes Químicos del Agua/toxicidad
20.
Fertil Steril ; 118(3): 494-503, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35842313

RESUMEN

OBJECTIVE: To study the consequences of specific genotype profiles of follicle-stimulating hormone receptor (FSHR) and luteinizing hormone choriogonadotropin receptor (LHCGR) on assisted reproductive technology outcomes when preimplantation genetic testing for aneuploidy is used for controlling the embryo ploidy status. The most common reported single-nucleotide polymorphisms in the amino acid position for the FSHR (N680S; N: asparagine, S: serine; [rs6166]) and the LHCGR (N312S variant; N: asparagine, S: serine [rs2293275]) were chosen for this study. DESIGN: Retrospective cohort study. SETTING: Private Fertility Clinic. PATIENT(S): All women aged 18-40 years undergoing their first assisted reproductive technology cycle with aneuploidy screening between 2006 and 2017 with body mass index of >18 and <40 kg/m2 were included. INTERVENTION(S): All patients received both recombinant follicle-stimulating hormone and human menopausal gonadotropin or low dose human chorionic gonadotropin. Genomic DNA was isolated from patients' blood. Genotyping of the FSHR and LHCGR polymorphisms was performed using TaqMan genotyping assays. Associations between both receptor genotypes and clinical outcomes were assessed using generalized regression and ANOVA. MAIN OUTCOMES MEASURE(S): Live birth rate was the primary outcome. Secondary outcomes included oocyte yield, mature oocytes, blastulation rate, usable blastocyst rate, and implantation rate. RESULT(S): A total of 1,183 patients met the inclusion criteria and generated reliable genotype results. The overall genotype frequencies in the study population for the FSHR gene were as follows: 21.7% homozygous for S in codon 680, 29.2% homozygous for N680, and 48.1% heterozygous (N680S). As for the LHCGR, 15.6% were homozygous for N312, 38.5% homozygous for S312 and 45.9% heterozygous (N312S). Our study population consisted of 53.8% non-Hispanic white; 6.1% Hispanic white; 4.1% Afro-American; 15.4% Asian; and 20.6% other or unknown. No significant association was found with any of the studied variables (oocyte yield, usable blastocyst rate, implantation rate, live birth) when genotypes were analyzed per receptor or in combination with one another. There was a statistically significant but clinically irrelevant difference in the rate of mature oocytes across different variant combinations. CONCLUSION(S): Our findings suggest that the presence of gonadotropin receptor polymorphisms in both FSHR N680S and LHCGR N312S are not associated with assisted reproductive technology outcomes; therefore, these variants should not be considered reproductive predictors.


Asunto(s)
Nacimiento Vivo , Receptores de HFE , Receptores de HL , Femenino , Humanos , Embarazo , Aneuploidia , Asparagina/genética , Gonadotropina Coriónica , Hormona Folículo Estimulante , Hormona Luteinizante , Menotropinas , Polimorfismo de Nucleótido Simple , Receptores de HFE/genética , Receptores de HL/genética , Técnicas Reproductivas Asistidas , Estudios Retrospectivos , Serina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA