Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.220
Filter
1.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39254333

ABSTRACT

There has been an alarming trend toward earlier puberty in girls, suggesting the influence of an environmental factor(s). As the reactivation of the reproductive axis during puberty is thought to be mediated by the hypothalamic neuropeptides kisspeptin and gonadotropin-releasing hormone (GnRH), we asked whether an environmental compound might activate the kisspeptin (KISS1R) or GnRH receptor (GnRHR). We used GnRHR or KISS1R-expressing HEK293 cells to screen the Tox21 10K compound library, a compendium of pharmaceuticals and environmental compounds, for GnRHR and KISS1R activation. Agonists were identified using Ca2+ flux and phosphorylated extracellularly regulated kinase (p-ERK) detection assays. Follow-up studies included measurement of genes known to be upregulated upon receptor activation using relevant murine or human cell lines and molecular docking simulation. Musk ambrette was identified as a KISS1R agonist, and treatment with musk ambrette led to increased expression of Gnrh1 in murine and human hypothalamic cells and expansion of GnRH neuronal area in developing zebrafish larvae. Molecular docking demonstrated that musk ambrette interacts with the His309, Gln122, and Gln123 residues of the KISS1R. A group of cholinergic agonists with structures similar to methacholine was identified as GnRHR agonists. When applied to murine gonadotrope cells, these agonists upregulated Fos, Jun, and/or Egr1. Molecular docking revealed a potential interaction between GnRHR and 5 agonists, with Asn305 constituting the most conservative GnRHR binding site. In summary, using a Tox21 10K compound library screen combined with cellular, molecular, and structural biology techniques, we have identified novel environmental agents that may activate the human KISS1R or GnRHR.


Subject(s)
Receptors, Kisspeptin-1 , Receptors, LHRH , Humans , Female , Animals , Receptors, Kisspeptin-1/metabolism , Receptors, Kisspeptin-1/genetics , Receptors, LHRH/metabolism , Receptors, LHRH/genetics , Mice , HEK293 Cells , Zebrafish , Gonadotropin-Releasing Hormone/metabolism , Puberty/drug effects , Hypothalamus/metabolism , Hypothalamus/drug effects , Molecular Docking Simulation , Sexual Maturation/drug effects , Sexual Maturation/physiology , Kisspeptins/metabolism , Kisspeptins/genetics , Environmental Pollutants/toxicity , Environmental Pollutants/pharmacology
2.
Bull Exp Biol Med ; 177(4): 436-441, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39264556

ABSTRACT

We studied the effect of a high-fat, high-carbohydrate diet (HFHCD) on basal testosterone levels in the blood and testosterone, its precursors, and expression of steroidogenic genes in the testes of rats treated with human chorionic gonadotropin (hCG, 10 IU/rat, subcutaneously, once), gonadotropin-releasing hormone receptor antagonist cetrorelix (75 µg/kg, subcutaneously, 3 days), and their combination. In HFHCD rats, no obvious signs of androgen deficiency were observed and the response of the testes to hCG stimulation was preserved. Unlike control rats (normal diet), the expression of the luteinizing hormone receptor gene in these rats did not change in response to hCG stimulation and cetrorelix administration; they also showed a paradoxical, more pronounced response to hCG administration under conditions of suppression of the gonadotropin secretion by cetrorelix. This suggests that the etiology and pathogenesis of obesity may have different effects on the hormonal status of the male reproductive system.


Subject(s)
Chorionic Gonadotropin , Gonadotropin-Releasing Hormone , Obesity , Testis , Testosterone , Male , Animals , Chorionic Gonadotropin/pharmacology , Obesity/metabolism , Obesity/drug therapy , Rats , Testosterone/blood , Testis/drug effects , Testis/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/analogs & derivatives , Receptors, LHRH/metabolism , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/genetics , Diet, High-Fat/adverse effects , Hormone Antagonists/pharmacology , Humans , Rats, Wistar
3.
Reprod Fertil Dev ; 362024 Sep.
Article in English | MEDLINE | ID: mdl-39283977

ABSTRACT

Context Understanding of central nervous system mechanisms related to age-related infertility remains limited. Fibril α-synuclein, distinct from its monomer form, is implicated in age-related diseases and propagates among neurons akin to prions. Aims We compared α-synuclein expression in gonadotropin-releasing hormone-expressing neurons (GnRH neurons) in the pre-optic area, arcuate nucleus, and median eminence of healthy heifers and aged cows to determine its role in age-related infertility. Methods We analysed mRNA and protein expression, along with fluorescent immunohistochemistry for GnRH and α-synuclein, followed by Congo red staining to detect amyloid deposits, and confocal microscopy. Key results Both mRNA and protein expressions of α-synuclein were confirmed by reverse transcription-polymerase chain reaction (RT-PCR) and western blots in bovine cortex, hippocampus, and anterior and posterior hypothalamus tissues. Significant differences in α-synuclein mRNA expression were observed in the cortex and hippocampus between young and old cows. Western blots showed five bands of α-synuclein, probably reflecting monomer, dimer, and oligomers, in the cortex, hippocampus, hypothalamus tissues, and there were significant differences in some bands between young and old cows. Bright-field and polarised light microscopy did not detect obvious amyloid deposition in aged hypothalami; however, higher-sensitive confocal microscopy unveiled strong positive signal of Congo red and α-synuclein in GnRH neurons in aged hypothalami. Additionally, α-synuclein expression was detected in immortalised GnRH neurons, GT1-7 cells. Conclusion Alpha-synuclein was expressed in GnRH neurons, and some differences were observed between young and old hypothalami. Implications Alpha-synuclein may play an important role in aging-related infertility.


Subject(s)
Aging , Gonadotropin-Releasing Hormone , Hypothalamus , Neurons , alpha-Synuclein , Animals , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Cattle , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Neurons/metabolism , Hypothalamus/metabolism , Female , Aging/metabolism , RNA, Messenger/metabolism
4.
BMC Res Notes ; 17(1): 260, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39267194

ABSTRACT

This study hypothesized that SCFA, acetate impacts positively on hypothalamic pyroptosis and its related abnormalities in experimentally induced PCOS rat model, possibly through NrF2/HIF1-α modulation. Eight-week-old female Wister rats were divided into groups (n = 5), namely control, PCOS, acetate and PCOS + acetate groups. Induction of PCOS was performed by administering 1 mg/kg body weight of letrozole for 21 days. After PCOS confirmation, the animals were treated with 200 mg/kg of acetate for 6 weeks. Rats with PCOS were characterized with insulin resistance, leptin resistance, increased plasma testosterone as well as degenerated ovarian follicles. There was also a significant increase in hypothalamic triglyceride level, triglyceride-glucose index, inflammatory biomarkers (SDF-1 and NF-kB) and caspase-6 as well as plasma LH and triglyceride. A decrease was observed in plasma adiponectin, GnRH, FSH, and hypothalamic GABA with severe inflammasome expression in PCOS rats. These were accompanied by decreased level of NrF2/HIF1-α, and the alterations were reversed when treated with acetate. Collectively, the present results suggest the therapeutic impact of acetate on hypothalamic pyroptosis and its related comorbidity in PCOS, a beneficial effect that is accompanied by modulation of NrF2/HIF1-α.


Subject(s)
Hypothalamus , Hypoxia-Inducible Factor 1, alpha Subunit , Polycystic Ovary Syndrome , Pyroptosis , Rats, Wistar , Polycystic Ovary Syndrome/chemically induced , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/pathology , Female , Animals , Hypothalamus/metabolism , Hypothalamus/drug effects , Hypothalamus/pathology , Pyroptosis/drug effects , Rats , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Resistance , NF-E2-Related Factor 2/metabolism , Disease Models, Animal , Letrozole/pharmacology , Triglycerides/blood , Triglycerides/metabolism , Luteinizing Hormone/blood , Follicle Stimulating Hormone/blood , Adiponectin/metabolism , Adiponectin/blood , Testosterone/blood , Leptin/blood , Leptin/metabolism , Gonadotropin-Releasing Hormone/metabolism , gamma-Aminobutyric Acid/metabolism
5.
Theriogenology ; 229: 202-213, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39217649

ABSTRACT

BACKGROUND: The hypothalamic-pituitary-gonadal (HPG) axis is pivotal in regulating reproductive functions, with gonadotropin-releasing hormone (GnRH) acting as a central regulator. Recently, polyamines have been shown to regulate the HPG axis, including GnRH expression and ovarian biology in old and adult rodents. The present study firstly highlights the age-specific variation in the polyamine and their corresponding biosynthetic enzymes in the ovary during aging, and further, the study focuses on the effect of polyamines, putrescine, and agmatine, in young female mice. METHOD AND RESULT: Immunofluorescence analysis revealed age-related differences in the expression of ornithine decarboxylase 1 (ODC1), spermine (SPM), and spermidine (SPD) in the ovaries, with adult mice exhibiting significantly higher expression levels compared to young and old mice. Likewise, qPCR analysis showed the mRNA levels of Odc1, Spermidine synthase (Srm), and Spermine synthase (Sms) show a significant increase in adult ovaries, which is then followed by a significant decline in old age. Histological examination demonstrated morphological alterations in the ovaries with age, including decreased follicle numbers and increased stromal cells in old mice. Furthermore, treatment with putrescine, a polyamine, in young mice resulted in larger ovaries and increased follicle numbers compared to controls. Additionally, serum levels of gonadotropin-releasing hormone (GnRH) and progesterone (P4) were measured, showing elevated levels in polyamine-treated mice. GnRH mRNA expression also increased significantly. Gene expression analysis revealed upregulation of genes associated with folliculogenesis such as Fshr, Bmp15, Gdf9, Amh, Star, Hsdb3, and Plaur in the ovaries and onset of puberty such as Tac2, and Kiss1, and a decrease in Mkrn3 in the hypothalamus of polyamine-treated mice. CONCLUSION: This study investigates the effect of polyamines in young immature female mice, shedding light on their role in upregulating GnRH, and enhancing folliculogenesis. Overall, these findings suggest that polyamines play a crucial role in ovarian aging and HPG axis regulation, offering potential therapeutics to reinstate fertility in reproductively challenged individuals.


Subject(s)
Gonadotropin-Releasing Hormone , Sexual Maturation , Animals , Female , Gonadotropin-Releasing Hormone/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Mice , Sexual Maturation/drug effects , Ovarian Follicle/drug effects , Ovarian Follicle/metabolism , Polyamines/metabolism , Aging , Ovary/drug effects , Ovary/metabolism , Gene Expression Regulation/drug effects
6.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39253941

ABSTRACT

Reproductive function in mammals depends on the ability of progesterone (P4) to suppress pulsatile gonadotrophin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion in a homeostatic-negative feedback loop. Previous research identified that cells upstream from GnRH neurons expressing the nuclear progesterone receptor (PGR) are required for P4-negative feedback. However, the identity of these cells and the mechanism by which they reduce GnRH/LH pulsatile secretion is unknown. We aimed to address the hypothesis that PGR expressed by a neural population in the arcuate nucleus recently identified as the GnRH pulse generator, cells expressing kisspeptin, neurokinin B, and dynorphin (KNDy cells), mediate P4-negative feedback. To achieve this, we used female mice with the PGR gene conditionally deleted from kisspeptin cells (KPRKO mice) and observed a substantial decrease in the percentage of KNDy neurons coexpressing PGR messenger RNA (mRNA) (11% in KPRKO mice vs 86% in wild-type [WT] mice). However, KPRKO mice did not display changes in the frequency or amplitude of LH pulses in diestrus or estrus, nor in the ability of exogenous P4 to blunt a postcastration increase in LH. Further, mRNA expression of arcuate kisspeptin and dynorphin, which are excitatory and inhibitory to GnRH secretion, respectively, remained unaltered in KPRKO mice compared to WT controls. Together, these findings show that the near-complete loss of PGR signaling from KNDy cells does not affect negative feedback regulation of GnRH pulse generation in mice, suggesting that feedback through this receptor can occur via a small number of KNDy cells or a yet unidentified cell population.


Subject(s)
Arcuate Nucleus of Hypothalamus , Feedback, Physiological , Gonadotropin-Releasing Hormone , Kisspeptins , Luteinizing Hormone , Mice, Knockout , Progesterone , Receptors, Progesterone , Animals , Female , Kisspeptins/metabolism , Kisspeptins/genetics , Receptors, Progesterone/metabolism , Receptors, Progesterone/genetics , Luteinizing Hormone/metabolism , Mice , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Arcuate Nucleus of Hypothalamus/metabolism , Progesterone/metabolism , Dynorphins/metabolism , Dynorphins/genetics , Neurons/metabolism , Neurokinin B/genetics , Neurokinin B/metabolism
7.
Reprod Domest Anim ; 59(9): e14722, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39295165

ABSTRACT

The objective of the study was to characterise the expression patterns of the two key components of cortisol action namely HSD11B1 (11-beta-hydroxysteroid dehydrogenase type 1) and NR3C1 (nuclear receptor subfamily 3, group C, member 1, also known as the glucocorticoid receptor) in superovulation induced bovine follicles during the periovulation and subsequent corpus luteum (CL) formation. Bovine ovaries containing preovulatory follicles or CL were timely defined during induced ovulation as follows: 0 h before GnRH (Gonadotropin-releasing hormone) application, and 4, 10, 20, 25 (follicles) and 60 h (early CL) after GnRH. The low mRNA expression of HSD11B1 and NR3C1 in the follicle group before the GnRH application increased significantly in the follicle group 20 h after GnRH and remained high afterward also in the early CL group. In contrast, the high NR3C1 mRNA decreased in follicles 25 h after GnRH (close to ovulation) and significantly increased again after ovulation (early CL). Our results indicated the involvement of HSD11B1 and NR3C1 as the two key components of cortisol action in the local mechanisms coordinating final follicle maturation, ovulation, follicular-luteal transition and CL development in the cow.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1 , Corpus Luteum , Gonadotropin-Releasing Hormone , Ovarian Follicle , Receptors, Glucocorticoid , Animals , Female , Cattle/physiology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Gonadotropin-Releasing Hormone/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , Ovulation Induction/veterinary , Ovulation/physiology , Gene Expression Regulation
8.
Int J Mol Sci ; 25(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39337542

ABSTRACT

Sexual maturation in goats is a dynamic process regulated precisely by the hypothalamic-pituitary-gonadal axis and is essential for reproduction. The hypothalamus plays a crucial role in this process and is the control center of the reproductive activity. It is significant to study the molecular mechanisms in the hypothalamus regulating sexual maturation in goats. We analyzed the serum hormone profiles and hypothalamic mRNA expression profiles of female goats during sexual development (1 day old (neonatal, D1, n = 5), 2 months old (prepuberty, M2, n = 5), 4 months old (sexual maturity, M4, n = 5), and 6 months old (breeding period, M6, n = 5)). The results indicated that from D1 to M6, serum hormone levels, including FSH, LH, progesterone, estradiol, IGF1, and leptin, exhibited an initial increase followed by a decline, peaking at M4. Furthermore, we identified a total of 508 differentially expressed genes in the hypothalamus, with a total of four distinct expression patterns. Nuclear receptor subfamily 1, group D, member 1 (NR1D1), glucagon-like peptide 1 receptor (GLP1R), and gonadotropin-releasing hormone 1 (GnRH-1) may contribute to hormone secretion, energy metabolism, and signal transduction during goat sexual maturation via circadian rhythm regulation, ECM receptor interactions, neuroactive ligand-receptor interactions, and Wnt signaling pathways. This investigation offers novel insights into the molecular mechanisms governing the hypothalamic regulation of goat sexual maturation.


Subject(s)
Goats , Hypothalamus , Sexual Maturation , Transcriptome , Animals , Goats/genetics , Goats/growth & development , Hypothalamus/metabolism , Sexual Maturation/genetics , Female , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Gene Expression Profiling , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism
9.
Endocrinology ; 165(10)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39248143

ABSTRACT

Luteinizing hormone (LH), a heterodimeric glycoprotein produced by pituitary gonadotrope cells, regulates gonadal function. Hypothalamic gonadotropin-releasing hormone (GnRH) stimulates LH synthesis and secretion. GnRH induces LHß subunit (Lhb) expression via the transcription factor, early growth response 1 (EGR1), acting on the Lhb promoter. In contrast, overexpression of zinc finger E-box binding homeobox 1 (ZEB1) represses LH production in mice, but the underlying mechanism was not previously elucidated. Here, we observed that ZEB1 inhibited GnRH-stimulated but not basal Lhb mRNA expression in homologous murine LßT2 cells. Moreover, ZEB1 blocked GnRH and/or EGR1 induction of murine Lhb but not human LHB promoter-reporter activity in these cells. Using chimeric reporters, we mapped the species-specific ZEB1 sensitivity to sequence differences, including in Z- and E-boxes, in the proximal Lhb/LHB promoters, immediately upstream of the transcription start sites. ZEB1 bound to the murine Lhb promoter with higher affinity than to the human LHB promoter in this region. To examine ZEB1's physiological role in LH synthesis, we characterized gonadotrope-specific Zeb1 knockout mice. Loss of ZEB1 in gonadotropes did not affect LH production or secretion. Collectively, the data suggest that ZEB1, when overexpressed, can inhibit GnRH/EGR1 induction of murine Lhb transcription but does not play a necessary role in LH synthesis in mice.


Subject(s)
Gonadotropin-Releasing Hormone , Luteinizing Hormone, beta Subunit , Luteinizing Hormone , Promoter Regions, Genetic , Zinc Finger E-box-Binding Homeobox 1 , Animals , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Mice , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/genetics , Luteinizing Hormone, beta Subunit/genetics , Luteinizing Hormone, beta Subunit/metabolism , Luteinizing Hormone/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Humans , Transcription, Genetic , Mice, Knockout , Cell Line , Gene Expression Regulation , Male
10.
J Clin Invest ; 134(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39087475

ABSTRACT

Kisspeptin is an essential neuropeptide sitting at the apex of the hypothalamo-pituitary-gonadal (HPG) endocrine axis to regulate gonadotropin-releasing hormone (GnRH) neurons and downstream reproductive hormones. Kisspeptin neurons integrate feedback from sex steroids facilitating regulation of the menstrual cycle and mediate the effects of metabolic stressors on the reproductive axis. In this issue of the JCI, Torres and colleagues describe another pathway for kisspeptin signaling in astrocytes to influence GnRH neuronal output. Astrocytes had kisspeptin receptors that activated canonical intracellular signaling pathways to constrain the magnitude of kisspeptin-induced GnRH neuronal stimulation. Additionally, the appositions between kisspeptin and GnRH neurons were dynamic during the ovarian cycle, with astrocyte kisspeptin signaling proposed as a putative modulator of this neuroplasticity. Importantly, astrocyte kisspeptin signaling also mediated susceptibility to metabolic stressors and the development of obesity-induced hypogonadism, underscoring the physiological and pathological importance of this pathway and revealing the importance of nonneuronal signaling in reproductive health.


Subject(s)
Astrocytes , Gonadotropin-Releasing Hormone , Kisspeptins , Neurons , Signal Transduction , Astrocytes/metabolism , Humans , Gonadotropin-Releasing Hormone/metabolism , Animals , Female , Neurons/metabolism , Kisspeptins/metabolism , Menstrual Cycle/metabolism , Reproduction
SELECTION OF CITATIONS
SEARCH DETAIL