Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 2.068
Filtrar
Más filtros

Publication year range
1.
Physiol Rev ; 100(2): 869-943, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31625459

RESUMEN

In humans, as in the other mammals, the neuroendocrine control of reproduction is ensured by the brain-pituitary gonadotropic axis. Multiple internal and environmental cues are integrated via brain neuronal networks, ultimately leading to the modulation of the activity of gonadotropin-releasing hormone (GnRH) neurons. The decapeptide GnRH is released into the hypothalamic-hypophysial portal blood system and stimulates the production of pituitary glycoprotein hormones, the two gonadotropins luteinizing hormone and follicle-stimulating hormone. A novel actor, the neuropeptide kisspeptin, acting upstream of GnRH, has attracted increasing attention in recent years. Other neuropeptides, such as gonadotropin-inhibiting hormone/RF-amide related peptide, and other members of the RF-amide peptide superfamily, as well as various nonpeptidic neuromediators such as dopamine and serotonin also provide a large panel of stimulatory or inhibitory regulators. This paper addresses the origin and evolution of the vertebrate gonadotropic axis. Brain-pituitary neuroendocrine axes are typical of vertebrates, the pituitary gland, mediator and amplifier of brain control on peripheral organs, being a vertebrate innovation. The paper reviews, from molecular and functional perspectives, the evolution across vertebrate radiation of some key actors of the vertebrate neuroendocrine control of reproduction and traces back their origin along the vertebrate lineage and in other metazoa before the emergence of vertebrates. A focus is given on how gene duplications, resulting from either local events or from whole genome duplication events, and followed by paralogous gene loss or conservation, might have shaped the evolutionary scenarios of current families of key actors of the gonadotropic axis.


Asunto(s)
Evolución Molecular , Duplicación de Gen , Genoma Humano , Gonadotropinas/genética , Gónadas/fisiología , Sistema Hipotálamo-Hipofisario/fisiología , Células Neuroendocrinas/fisiología , Reproducción/genética , Animales , Gonadotropinas/metabolismo , Gónadas/metabolismo , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Células Neuroendocrinas/metabolismo , Filogenia , Especificidad de la Especie
2.
BMC Biol ; 22(1): 104, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702712

RESUMEN

BACKGROUND: Gonadotropin precisely controls mammalian reproductive activities. Systematic analysis of the mechanisms by which epigenetic modifications regulate the synthesis and secretion of gonadotropin can be useful for more precise regulation of the animal reproductive process. Previous studies have identified many differential m6A modifications in the GnRH-treated adenohypophysis. However, the molecular mechanism by which m6A modification regulates gonadotropin synthesis and secretion remains unclear. RESULTS: Herein, it was found that GnRH can promote gonadotropin synthesis and secretion by promoting the expression of FTO. Highly expressed FTO binds to Foxp2 mRNA in the nucleus, exerting a demethylation function and reducing m6A modification. After Foxp2 mRNA exits the nucleus, the lack of m6A modification prevents YTHDF3 from binding to it, resulting in increased stability and upregulation of Foxp2 mRNA expression, which activates the cAMP/PKA signaling pathway to promote gonadotropin synthesis and secretion. CONCLUSIONS: Overall, the study reveals the molecular mechanism of GnRH regulating the gonadotropin synthesis and secretion through FTO-mediated m6A modification. The results of this study allow systematic interpretation of the regulatory mechanism of gonadotropin synthesis and secretion in the pituitary at the epigenetic level and provide a theoretical basis for the application of reproductive hormones in the regulation of animal artificial reproduction.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Hormona Liberadora de Gonadotropina , Animales , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/genética , Gonadotropinas/metabolismo , Metilación de ARN , ARN Mensajero/metabolismo , ARN Mensajero/genética , Ratas
3.
Hum Reprod ; 39(10): 2331-2340, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39180767

RESUMEN

STUDY QUESTION: Does medroxyprogesterone acetate (MPA) exposure in progestin-primed ovarian stimulation (PPOS) cycles cause molecular perturbations in the steroidogenic function and gonadotropin responsiveness of the granulosa cells? SUMMARY ANSWER: PPOS cycles are identical to traditional GnRH antagonist cycles not only for clinical IVF characteristics but also for gonadotropin receptor expression, response to gonadotropins, and steroidogenic function at the molecular level. WHAT IS KNOWN ALREADY: PPOS is increasingly used as an alternative to GnRH antagonists due to the inhibitory effect of progesterone on LH release by reducing GnRH pulsatility at the hypothalamic level. Although a growing body of evidence from clinical studies did not indicate significant differences between PPOS and antagonist protocols for IVF cycle characteristics and obstetrical outcomes, it is still unknown whether exposure of the antral follicle cohort to progesterone or its synthetic derivatives during ovarian stimulation causes any subtle molecular aberrations in terms of steroidogenesis and gonadotropin responsiveness. To address this issue, detailed comparative molecular analyses were conducted in the luteinized mural granulosa cells (GCs) obtained from normal responding IVF patients undergoing PPOS and antagonist cycles. STUDY DESIGN, SIZE, DURATION: A clinical translational research study was conducted with IVF patients. PARTICIPANTS/MATERIALS, SETTING, METHODS: This study included 55 normal responding IVF patients who underwent ovarian stimulation with either PPOS using MPA (5 mg twice daily) or GnRH antagonist cetrorelix acetate. Recombinant forms of FSH and hCG were used for ovarian stimulation and ovulation triggering, respectively. Luteinized mural GCs obtained during the oocyte retrieval procedure were used for the experiments. Cell culture, quantitative real-time PCR, immunoblotting, confocal time-lapse live cell imaging, and hormone assays were used. MAIN RESULTS AND THE ROLE OF CHANCE: Demographic and IVF cycle characteristics of the patients undergoing ovarian stimulation with PPOS and GnRH antagonist were similar, including ovarian response, mature oocyte yield, and fertilization rates. Molecular analyses revealed that the expression of the enzymes involved in sex-steroid synthesis (StAR, SCC, 3ß-HSD, 17ß-HSD, aromatase) and the uptake/storage/utilization of cholesterol (LDL receptor, Hormone-sensitive lipase, hydroxy-methyl glutaryl Co-enzyme-A reductase, and Sterol O-acyltransferase1) in the GCs of the PPOS cycles were comparable to those of the antagonist cycles. The expression of the receptors for gonadotropins, estrogen, and progesterone hormones was also similar. Basal and hCG-induced increases in 3ß-HSD expression and progesterone production and basal and FSH-induced increases in aromatase expression and E2 output of the GCs from PPOS patients did not exhibit any meaningful differences when compared with GCs from antagonist cycles. Furthermore, basal and hCG-induced up-regulation in the LDL receptor expression and cholesterol uptake did not differ between the groups. Confocal imaging also revealed similar patterns of expression for the steroidogenic enzymes and their co-localization with mitochondria. Lastly, the expression of the other important genes regulating cumulus expansion, ovulation, and luteal function [Relaxin, ADAMTS-1, and epidermal growth factor (EGF)-like growth factor amphiregulin] in the GCs of the PPOS and antagonist cycles were similar. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Caution should be exercised when interpreting our data which was derived from normally responding patients whose ovulation was triggered with hCG. It is unclear whether the molecular parameters assessed vary according to infertility etiologies, magnitude of ovarian response, mode of trigger, and any other underlying ovarian pathologies or systemic diseases. MPA was the progestin used for PPOS and whether these findings can be generalized to other progestins is unknown. WIDER IMPLICATIONS OF THE FINDINGS: This study provides reassuring molecular evidence that exposure of antral follicle cohorts to MPA during the follicular growth phase does not have any detrimental effects on steroidogenic, ovulatory, and luteal functions when compared with GnRH antagonist cycles. STUDY FUNDING/COMPETING INTEREST(S): This study was funded by the School of Medicine, the Graduate School of Health Sciences of Koc University and Koç University Research Center for Translational Medicine (KUTTAM), and equally funded by the Republic of Turkey Ministry of Development Research Infrastructure Support Program. All authors declare no conflict of interest. TRIAL REGISTRATION NUMBER: N/A.


Asunto(s)
Células de la Granulosa , Acetato de Medroxiprogesterona , Folículo Ovárico , Inducción de la Ovulación , Femenino , Humanos , Acetato de Medroxiprogesterona/farmacología , Inducción de la Ovulación/métodos , Células de la Granulosa/metabolismo , Células de la Granulosa/efectos de los fármacos , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Adulto , Fertilización In Vitro/métodos , Progestinas/farmacología , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/metabolismo
4.
Cell Commun Signal ; 22(1): 398, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143495

RESUMEN

Ovarian granulosa cells are essential to gonadotrophin-regulated estrogen production, female cycle maintenance and fertility. The epithelial Na+ channel (ENaC) is associated with female fertility; however, whether and how it plays a role in ovarian cell function(s) remained unexplored. Here, we report patch-clamp and Na+ imaging detection of ENaC expression and channel activity in both human and mouse ovarian granulosa cells, which are promoted by pituitary gonadotrophins, follicle stimulating hormone (FSH) or luteinizing hormone (LH). Cre-recombinase- and CRISPR-Cas9-based granulosa-specific knockout of ENaC α subunit (Scnn1a) in mice resulted in failed estrogen elevation at early estrus, reduced number of corpus luteum, abnormally extended estrus phase, reduced litter size and subfertility in adult female mice. Further analysis using technologies including RNA sequencing and Ca2+ imaging revealed that pharmacological inhibition, shRNA-based knockdown or the knockout of ENaC diminished spontaneous or stimulated Ca2+ oscillations, lowered the capacity of intracellular Ca2+ stores and impaired FSH/LH-stimulated transcriptome changes for estrogen production in mouse and/or human granulosa cells. Together, these results have revealed a previously undefined role of ENaC in modulating gonadotrophin signaling in granulosa cells for estrogen homeostasis and thus female fertility.


Asunto(s)
Calcio , Canales Epiteliales de Sodio , Estrógenos , Fertilidad , Células de la Granulosa , Homeostasis , Femenino , Animales , Células de la Granulosa/metabolismo , Canales Epiteliales de Sodio/metabolismo , Canales Epiteliales de Sodio/genética , Humanos , Estrógenos/metabolismo , Ratones , Fertilidad/genética , Calcio/metabolismo , Gonadotropinas/metabolismo , Transducción de Señal , Ratones Noqueados , Señalización del Calcio
5.
Neuroendocrinology ; 114(3): 234-249, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37899035

RESUMEN

INTRODUCTION: Apelin is an endogenous peptide, whose expression has been shown in the hypothalamus, pituitary, and ovary; furthermore, it is also called a neuropeptide, binding to apelin receptor (APJ) for various functions. It has been suggested that the hypothalamus, pituitary, and ovarian (HPO) axis is tightly regulated and factors and functions of the HPO axis can be modulated during the estrous cycle to influence reproductive status. To the best of our knowledge, the status of apelin and its receptor, APJ has not been investigated in the HPO axis during the estrous cycle. METHODS: To explore the expression of apelin and APJ in the HPO axis of mice during the estrous cycle, mice were divided into four groups: proestrus (Pro), estrus (Est), metestrus (Met), and diestrus (Di), and apelin and APJ were checked. Further, to explore the role of apelin in gonadotropin secretion, an in vitro study of the pituitary was performed at the Pro and Est stages. RESULT: The expression apelin and APJ in the hypothalamus showed elevation during the estrous cycle of postovulatory phases, Met, and Di. The immunolocalization of apelin and APJ in the anterior pituitary showed more abundance in the Est and Di. Our in vitro results showed that gonadotropin-releasing hormone agonist stimulated luteinizing hormone secretion was suppressed by the apelin 13 peptide from the pituitary of Pro and Est phases. This suggests an inhibitory role of apelin on gonadotropin secretion. The ovary also showed conspicuous changes in the presence of apelin and APJ during the estrous cycle. The expression of apelin and APJ coincides with folliculogenesis and corpus luteum formation and the expression of the apelin system in the different cell types of the ovary suggests its cell-specific role. Previous studies also showed that apelin has a stimulatory role in ovarian steroid secretion, proliferation, and corpus luteum. CONCLUSION: Overall our results showed that the apelin system changes along the HPO axis during the estrous cycle and might have an inhibitory at level of hypothalamus and pituitary and a stimulatory role at ovarian level.


Asunto(s)
Ovario , Enfermedades de la Hipófisis , Animales , Femenino , Ratones , Apelina/metabolismo , Receptores de Apelina/metabolismo , Ciclo Estral , Gonadotropinas/metabolismo , Ovario/metabolismo
6.
Br J Clin Pharmacol ; 90(10): 2387-2397, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39187392

RESUMEN

Adolescent transgender medicine is a growing clinical field. Gender-affirming medications for transgender youth may include gonadotropin-releasing hormone (GnRH) agonists, gender-affirming hormones or both. To evaluate the potential effects of GnRH agonists (puberty suppression) on pharmacokinetic processes for transgender youth, we searched PubMed from inception to May 2024 for publications on the effects of GnRH agonists on drug absorption, distribution, metabolism or excretion for transgender adolescents or effects on hormones (including gonadotropins, adrenal androgens, sex steroids) that are associated with changes in drug metabolism during puberty in the general adolescent population. No publications discussed the effects of GnRH agonist treatment on pharmacokinetic processes for adolescent transgender people. Sixteen publications observed marked decreases in gonadotropins and sex steroids for both adolescent transgender men and adolescent transgender women and slight effects on adrenal androgens. During GnRH agonist treatment, changes in body composition and body shape were greater for adolescent transgender people than for cisgender adolescent people. Further research is needed to better understand the effects of GnRH agonists on drug metabolism and other pharmacokinetic processes for transgender adolescents receiving GnRH agonists and other gender-affirming medications.


Asunto(s)
Hormona Liberadora de Gonadotropina , Personas Transgénero , Humanos , Adolescente , Hormona Liberadora de Gonadotropina/agonistas , Masculino , Femenino , Hormonas Esteroides Gonadales , Andrógenos/farmacocinética , Gonadotropinas/metabolismo , Farmacología Clínica/métodos
7.
Gen Comp Endocrinol ; 347: 114425, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38101488

RESUMEN

The Pacific halibut (Hippoglossus stenolepis) is a large migratory demersal flatfish species that occupies a top trophic role in the North Pacific Ocean and Bering Sea ecosystems, where it also supports various fisheries. As a first attempt to characterize the endocrine mechanisms driving sexual maturation in this important species, we collected pituitary, ovarian and blood samples from Pacific halibut females captured in the wild that were classified histologically into various female developmental stages. We conducted gene expression analyses of gonadotropin beta subunits in the pituitary and observed that mRNA expression levels of fshb gradually increased throughout vitellogenesis, remained elevated until before ovulation and declined after spawning. In contrast, the mRNA expression levels of lhb markedly increased during oocyte maturation and remained elevated until after spawning. Ovarian mRNA expression levels of the gonadotropin receptor genes fshr and lhr peaked during oocyte maturation and before spawning, respectively, immediately following the developmental stage at which pituitary fshb and lhb mRNA expression first reached maximum levels. The ovarian gene expression patterns of steroidogenic enzyme genes cyp19a1 and hsd20b2 paralleled those of fshr and lhr, respectively. Testosterone and 17ß-estradiol (E2) plasma levels increased concomitantly with fshr and cyp19a1 mRNA expression levels, and vitellogenin plasma levels increased throughout vitellogenesis and reached maximum levels prior to spawning. These results are consistent with the notion that in female Pacific halibut, as in other teleosts, vitellogenesis and oocyte maturation and ovulation are likely under the control of pituitary gonadotropic hormones Fsh and Lh, respectively.


Asunto(s)
Lenguado , Animales , Femenino , Lenguado/genética , Lenguado/metabolismo , Ecosistema , Gonadotropinas Hipofisarias/metabolismo , Gonadotropinas/genética , Gonadotropinas/metabolismo , ARN Mensajero/genética
8.
Gen Comp Endocrinol ; 350: 114465, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38336122

RESUMEN

We compared the endocrine status of the pituitary-gonad axis of wild and captive-reared greater amberjack (Seriola dumerili) during the reproductive cycle (April - July), reporting on the expression and release of the two gonadotropins for the first time in the Mediterranean Sea. Ovaries from wild females were characterized histologically as DEVELOPING in early May and SPAWNING capable in late May-July, the latter having a 3 to 4-fold higher gonadosomatic index (GSI). SPAWNING capable wild females exhibited an increase in pituitary follicle stimulating hormone (Fsh) content, plasma testosterone (T) and 17,20ß-dihydroxy-4-pregnen-3-one (17,20ß-P), while almost a 10-fold increase was observed in pituitary luteinizing hormone (Lh) content. An increasing trend of plasma 17ß-estradiol (E2) was also recorded between the two reproductive stages in wild females. Captive-reared females sampled during the reproductive cycle exhibited two additional reproductive categories, with REGRESSED females having extensive follicular atresia and fish in the REGENERATING stage having only primary oocytes in their ovaries. Pituitary content of Fsh and Lh, fshb and lhb expression and plasma levels of Fsh and Lh remained unchanged among the four reproductive stages in captive females, in contrast with plasma E2 and T that decreased in the REGENERATING stage, and 17,20ß-P which increased after the DEVELOPING stage. In general, no significant hormonal differences were recorded between captive-reared and wild DEVELOPING females, in contrast to SPAWNING capable females, where pituitary Lh content, plasma Fsh and T were found to be lower in females in captivity. Overall, the captive females lagged behind in reproductive development compared to the wild ones and this was perhaps related to the multiple handling of the sea cages where all the sampled fish were maintained. Between wild males in the DEVELOPING and SPAWNING capable stages, pituitary Lh content, plasma T and 17,20ß-P, and GSI exhibited 3 to 4-fold increases, while an increasing trend of pituitary Fsh content, lhb expression levels and plasma 11-ketotestosterone (11-KT) was also observed, and an opposite trend was observed in plasma Lh. Captive males were allocated to one more category, with REGRESSED individuals having no spermatogenic capacity. During the SPAWNING capable phase, almost all measured parameters were lower in captive males compared to wild ones. More importantly, captive males showed significant differences from their wild counterparts throughout the reproductive season, starting already from the DEVELOPING stage. Therefore, it appears that captivity already exerted negative effects in males prior to the onset of the study and the multiple handling of the cage where sampled fish were reared. Overall, the present study demonstrated that female greater amberjack do undergo full vitellogenesis in captivity, albeit with some dysfunctions that may be related to the husbandry of the experiment, while males, on the other hand, may be more seriously affected by captivity even before the onset of the study.


Asunto(s)
Atresia Folicular , Perciformes , Animales , Masculino , Femenino , Gonadotropinas/metabolismo , Hormona Luteinizante/metabolismo , Reproducción , Hormona Folículo Estimulante/metabolismo , Perciformes/metabolismo , Hipófisis/metabolismo , Peces/metabolismo
9.
Gen Comp Endocrinol ; 350: 114477, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38387532

RESUMEN

Gonadotropin-inhibitory hormone (GnIH) was the first reported hypothalamic neuropeptide inhibiting reproduction in vertebrates. Since its discovery in the quail brain, its orthologs have been identified in a variety of vertebrate species and even protochordates. Depending on the species, the GnIH precursor polypeptides comprise two, three or four mature peptides of the RFamide family. It has been well documented that GnIH inhibits reproduction at the brain-pituitary-gonadal levels and participates in metabolism, stress response, and social behaviors in birds and mammals. However, most studies in fish have mainly been focused on the physiological roles of GnIH in the control of reproduction and results obtained are in some cases conflicting, leaving aside its potential roles in the regulation of other functions. In this manuscript we summarize the information available in fish with respect to the structural diversity of GnIH peptides and functional roles of GnIH in reproduction and other physiological processes. We also highlight the molecular mechanisms of GnIH actions on target cells and possible interactions with other neuroendocrine factors.


Asunto(s)
Gonadotropinas , Hormonas Hipotalámicas , Animales , Gonadotropinas/metabolismo , Vertebrados/metabolismo , Péptidos/metabolismo , Hipotálamo/metabolismo , Reproducción/fisiología , Peces/metabolismo , Mamíferos/metabolismo , Hormonas Hipotalámicas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo
10.
Gen Comp Endocrinol ; 357: 114593, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39047797

RESUMEN

Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) control antral follicular growth by regulating several processes, such as the synthesis of hormones and signaling molecules, proliferation, survival, apoptosis, luteinization, and ovulation. To exert these effects, gonadotropins bind to their respective Gs protein-coupled receptors, activating the protein kinase A (PKA) pathway or recruiting Gq proteins to activate protein kinase C (PKC) signaling. Although the action mechanism of FSH and LH is clear, recently, it has been shown that both gonadotropins promote the synthesis of sphingosine-1-phosphate (S1P) in granulosa and theca cells through the activation of sphingosine kinase 1. Moreover, the inhibition of SPHKs reduces S1P synthesis, cell viability, and the proliferation of follicular cells in response to gonadotropins, and the addition of S1P to the culture medium increases the proliferation of granulosa and theca cells without apparent effects on sexual steroid synthesis. Therefore, we consider that S1P is a crucial signaling molecule that complements the canonical gonadotropin pathway to promote the proliferation and viability of granulosa and theca cells.


Asunto(s)
Gonadotropinas , Lisofosfolípidos , Folículo Ovárico , Esfingosina , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacología , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Femenino , Animales , Humanos , Gonadotropinas/metabolismo , Folículo Ovárico/metabolismo , Folículo Ovárico/efectos de los fármacos , Hormona Luteinizante/metabolismo , Hormona Folículo Estimulante/metabolismo , Hormona Folículo Estimulante/farmacología , Transducción de Señal/efectos de los fármacos , Células de la Granulosa/metabolismo , Células de la Granulosa/efectos de los fármacos
11.
J Reprod Dev ; 70(2): 115-122, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38346724

RESUMEN

The NR4A nuclear receptor family (NR4As), encompassing NR4A1, NR4A2, and NR4A3, exerts pivotal roles in cellular processes through intricate expression patterns and interactions. Despite the influence of some NR4As on anterior pituitary functions regulated by the hypothalamus, their physiological expression patterns remain unclear. In our prior work, we demonstrated the specific upregulation of NR4A3 in the rat anterior pituitary gland during the proestrus afternoon, coinciding with a gonadotropin surge. In this study, we investigated changes in pituitary Nr4a gene expression throughout the estrous cycle in rats and a gonadotropin surge-induced model. Nr4a1 and Nr4a2 gene expression significantly increased during proestrus, aligning with previous observations for Nr4a3. Furthermore, prolactin gene expression increased sequentially with rising Nr4a gene expression, while thyroid-stimulating hormone beta gene expression remained stable. Immunohistochemistry revealed a widespread and differential distribution of NR4A proteins in the anterior pituitary, with NR4A1 and NR4A3 being particularly abundant in thyrotrophs, and NR4A2 in gonadotrophs. In estrogen-treated ovariectomized rats, elevated luteinizing hormone secretion corresponded to markedly upregulated expression of Nr4a1, Nr4a2, and Nr4a3. In gonadotroph and somatomammotroph cell lines, gonadotropin- and thyrotropin-releasing hormones transiently and dose-dependently increased the expression of Nr4a genes. These findings suggest that hypothalamic hormone secretion during proestrus may induce the parallel expression of pituitary Nr4a genes, potentially influencing the pituitary gene expression program related to endocrine functions before and after ovulation.


Asunto(s)
Adenohipófisis , Hipófisis , Femenino , Ratas , Animales , Proestro/fisiología , Hipófisis/metabolismo , Adenohipófisis/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/metabolismo
12.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33500349

RESUMEN

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.


Asunto(s)
Dinorfinas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/metabolismo , Kisspeptinas/metabolismo , Neuroquinina B/metabolismo , Neuronas/metabolismo , Organogénesis , Folículo Ovárico/crecimiento & desarrollo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Aromatasa/genética , Aromatasa/metabolismo , Retroalimentación Fisiológica , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Integrasas/metabolismo , Hormona Luteinizante/sangre , Tamaño de los Órganos , Folículo Ovárico/metabolismo , Hipófisis/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de HL/genética , Receptores de HL/metabolismo , Receptores LHRH/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-39089445

RESUMEN

Temperature is a preeminent factor in the regulation of fish reproduction and hinders gonadal development beyond a specific threshold. To comprehend the molecular mechanism responsible for reproductive suppression at different temperature, expression of the genes encoding kisspeptin (kiss2), gonadotropin-releasing hormone (gnrh1) and their receptors (gpr54, gnrh1r) in the brain, and the gonadotropin (GTH) subunits (fshb and lhb) in the pituitary were studied in juvenile Nile tilapia (Oreochromis niloticus) along with gonadal histology. Fish were acclimatized to three distinct temperatures, including 31 °C, 34 °C and 37 °C for 14 days. The mRNA levels of kiss2, gpr54, gnrh1, and gnrh1r were significantly decreased at 37 °C compared to 31 °C and 34 °C in the both sexes. In parallel, the expression level of fshb in the both sexes and lhb in the female were significantly lower at 37 °C in the pituitary. Histologically, the gonads of both sexes had normal growth of gametes at control temperature (31 °C), whereas the spermatogenesis and oocyte maturation were slowed down and atretic oocytes were found in the ovary at 37 °C acclimation temperature. Taken together, the results imply that elevated temperature beyond the specific threshold may have a negative impact on reproduction by suppressing the gene expressions of kisspeptin/GnRH1/GTH system and eventually restrains normal growth and maturation of gametes in the both sexes of Nile tilapia.


Asunto(s)
Cíclidos , Hormona Liberadora de Gonadotropina , Gónadas , Kisspeptinas , Animales , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Cíclidos/genética , Cíclidos/crecimiento & desarrollo , Cíclidos/metabolismo , Femenino , Masculino , Gónadas/metabolismo , Gónadas/crecimiento & desarrollo , Temperatura , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Hipófisis/metabolismo , Ovario/metabolismo , Ovario/crecimiento & desarrollo , Gonadotropinas/metabolismo , Regulación del Desarrollo de la Expresión Génica
14.
Int J Mol Sci ; 25(14)2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-39062762

RESUMEN

Female fertility depends on the ovarian reserve of follicles, which is determined at birth. Primordial follicle development and oocyte maturation are regulated by multiple factors and pathways and classified into gonadotropin-independent and gonadotropin-dependent phases, according to the response to gonadotropins. Folliculogenesis has always been considered to be gonadotropin-dependent only from the antral stage, but evidence from the literature highlights the role of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) during early folliculogenesis with a potential role in the progression of the pool of primordial follicles. Hormonal and molecular pathway alterations during the very earliest stages of folliculogenesis may be the root cause of anovulation in polycystic ovary syndrome (PCOS) and in PCOS-like phenotypes related to antiepileptic treatment. Excessive induction of primordial follicle activation can also lead to premature ovarian insufficiency (POI), a condition characterized by menopause in women before 40 years of age. Future treatments aiming to suppress initial recruitment or prevent the growth of resting follicles could help in prolonging female fertility, especially in women with PCOS or POI. This review will briefly introduce the impact of gonadotropins on early folliculogenesis. We will discuss the influence of LH on ovarian reserve and its potential role in PCOS and POI infertility.


Asunto(s)
Gonadotropinas , Folículo Ovárico , Síndrome del Ovario Poliquístico , Insuficiencia Ovárica Primaria , Animales , Femenino , Humanos , Hormona Folículo Estimulante/metabolismo , Gonadotropinas/metabolismo , Hormona Luteinizante/metabolismo , Folículo Ovárico/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/fisiopatología , Insuficiencia Ovárica Primaria/metabolismo , Insuficiencia Ovárica Primaria/etiología , Insuficiencia Ovárica Primaria/patología
15.
Front Neuroendocrinol ; 64: 100954, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34757092

RESUMEN

The social environment changes circulating hormone levels and expression of social behavior in animals. Social information is perceived by sensory systems, leading to cellular and molecular changes through neural processes. Peripheral reproductive hormone levels are regulated by activity in the hypothalamic-pituitary-gonadal (HPG) axis. Until the end of the last century, the neurochemical systems that convey social information to the HPG axis were not well understood. Gonadotropin-inhibitory hormone (GnIH) was the first hypothalamic neuropeptide shown to inhibit gonadotropin release, in 2000. GnIH is now regarded as a negative upstream regulator of the HPG axis, and it is becoming increasingly evident that it responds to social cues. In addition to controlling reproductive physiology, GnIH seems to modulate the reproductive behavior of animals. Here, we review studies investigating how GnIH neurons respond to social information and describe the mechanisms through which GnIH regulates social behavior.


Asunto(s)
Hormonas Hipotalámicas , Animales , Gonadotropinas/metabolismo , Hormonas Hipotalámicas/metabolismo , Hormonas Hipotalámicas/farmacología , Hipotálamo/metabolismo , Interacción Social , Vertebrados/metabolismo
16.
Front Neuroendocrinol ; 64: 100953, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34757094

RESUMEN

Under stressful condition, reproductive function is impaired due to the activation of various components of the hypothalamic-pituitaryadrenal (HPA) axis, which can suppress the activity of the hypothalamic-pituitary-gonadal (HPG) axis at multiple levels. A hypothalamic neuropeptide, gonadotropin-inhibitory hormone (GnIH) is a key negative regulator of reproduction that governs the HPG axis. Converging lines of evidence have suggested that different stress types and their duration, such as physical or psychological, and acute or chronic, can modulate the GnIH system. To clarify the sensitivity and reactivity of the GnIH system in response to stress, we summarize and critically review the available studies that investigated the effects of various stressors, such as restraint, nutritional/metabolic and social stress, on GnIH expression and/or its neuronal activity leading to altered HPG action. In this review, we focus on GnIH as the potential novel mediator responsible for stress-induced reproductive dysfunction.


Asunto(s)
Hormonas Hipotalámicas , Neuropéptidos , Gonadotropinas/metabolismo , Hormonas Hipotalámicas/metabolismo , Hormonas Hipotalámicas/farmacología , Hipotálamo/metabolismo , Neuropéptidos/metabolismo , Reproducción/fisiología
17.
Front Neuroendocrinol ; 65: 100991, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35227766

RESUMEN

This paper intends to apprise the reader regarding the existing knowledge on the neuroanatomical distribution of GnIH-like peptides in in fish and amphibians in both the adult stage and during ontogenesis. The neuroanatomical distribution of GnIH-like neuropeptides appears quite different in the studied species, irrespective of the evolutionary closeness. The topology of the olfactory bulbs can affect the distribution of neurons producing the GnIH-like peptides, with a tendency to show a more extended distribution into the brains with pedunculate olfactory bulbs. Therefore, the variability of the GnIH-like system could also reflect specific adaptations rather than evolutionary patterns. The onset of GnIH expression was detected very early during development suggesting its precocious roles, and the neuroanatomical distribution of GnIH-like elements showed a generally increasing trend. This review highlights some critical technical aspects and the need to increase the number of species to be studied to obtain a complete neuroanatomical picture of the GnIH-like system.


Asunto(s)
Hormonas Hipotalámicas , Neuropéptidos , Anfibios/metabolismo , Animales , Encéfalo/metabolismo , Gonadotropinas/metabolismo , Hormonas Hipotalámicas/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo
18.
Hum Mol Genet ; 30(10): 923-938, 2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-33856019

RESUMEN

Women heterozygous for an expansion of CGG repeats in the 5'UTR of FMR1 risk developing fragile X-associated primary ovarian insufficiency (FXPOI) and/or tremor and ataxia syndrome (FXTAS). We show that expanded CGGs, independent of FMR1, are sufficient to drive ovarian insufficiency and that expression of CGG-containing mRNAs alone or in conjunction with a polyglycine-containing peptide translated from these RNAs contribute to dysfunction. Heterozygous females from two mouse lines expressing either CGG RNA-only (RNA-only) or CGG RNA and the polyglycine product FMRpolyG (FMRpolyG+RNA) were used to assess ovarian function in aging animals. The expression of FMRpolyG+RNA led to early cessation of breeding, ovulation and transcriptomic changes affecting cholesterol and steroid hormone biosynthesis. Females expressing CGG RNA-only did not exhibit decreased progeny during natural breeding, but their ovarian transcriptomes were enriched for alterations in cholesterol and lipid biosynthesis. The enrichment of CGG RNA-only ovaries for differentially expressed genes related to cholesterol processing provided a link to the ovarian cysts observed in both CGG-expressing lines. Early changes in transcriptome profiles led us to measure ovarian function in prepubertal females that revealed deficiencies in ovulatory responses to gonadotropins. These include impairments in cumulus expansion and resumption of oocyte meiosis, as well as reduced ovulated oocyte number. Cumulatively, we demonstrated the sufficiency of ectopically expressed CGG repeats to lead to ovarian insufficiency and that co-expression of CGG-RNA and FMRpolyG lead to premature cessation of breeding. However, the expression of CGG RNA-alone was sufficient to lead to ovarian dysfunction by impairing responses to hormonal stimulation.


Asunto(s)
Ataxia/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Insuficiencia Ovárica Primaria/genética , Transcriptoma/genética , Temblor/genética , Animales , Ataxia/patología , Modelos Animales de Enfermedad , Expresión Génica Ectópica/genética , Femenino , Síndrome del Cromosoma X Frágil/patología , Gonadotropinas/metabolismo , Humanos , Ratones , Oocitos/crecimiento & desarrollo , Péptidos/genética , Insuficiencia Ovárica Primaria/patología , Temblor/patología , Expansión de Repetición de Trinucleótido/genética
19.
Development ; 147(22)2020 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-33060133

RESUMEN

Anti-Müllerian hormone (Amh) plays an important role in gonadal function. Amh deficiency causes severe gonadal dysgenesis and dysfunction in zebrafish, with gonadal hypertrophy in both sexes. However, its mechanism of action remains unknown. Intriguingly, the Amh cognate type II receptor (Amhr2) is missing in the zebrafish genome, in sharp contrast to other species. Using a series of zebrafish mutants (amh, fshb, fshr and lhcgr), we provided unequivocal evidence for actions of Amh, via modulation of gonadotropin signaling, on both germ cell proliferation and differentiation. The gonadal hypertrophy in amh mutants was abolished in the absence of Fshr in females or Fshr/Lhcgr in males. Furthermore, we demonstrated that knockout of bmpr2a, but not bmpr2b, phenocopied all phenotypes of the amh mutant in both sexes, including gonadal hypertrophy, hyperproliferation of germ cells, retarded gametogenesis and reduced fshb expression. In summary, the present study provided comprehensive genetic evidence for an intimate interaction of gonadotropin and Amh pathways in gonadal homeostasis and gametogenesis and for Bmpr2a as the possible missing link for Amh signaling in zebrafish.


Asunto(s)
Hormona Antimülleriana/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Gametogénesis/fisiología , Células Germinativas/metabolismo , Gonadotropinas/metabolismo , Transducción de Señal/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Hormona Antimülleriana/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Femenino , Células Germinativas/citología , Gonadotropinas/genética , Masculino , Pez Cebra/genética , Proteínas de Pez Cebra/genética
20.
Biol Reprod ; 109(6): 892-903, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-37698264

RESUMEN

Perinatal nutrition modulates the hypothalamic neurocircuitries controlling GnRH release, thus programming pubertal maturation in female mammals. Objectives of experiments reported here were to test the hypotheses that prenatal nutrition during mid- to late gestation interacts with postnatal nutrition during the juvenile period in heifer offspring to alter expression of leptin receptor (LepR) variants (ObRa, ObRb, ObRc, ObRt), and lipoprotein transporter molecules (LRP1 and 2) in the choroid plexus, leptin transport across the blood-brain barrier, and hypothalamic-hypophyseal responsiveness to exogenous ovine leptin (oleptin) during fasting. Nutritional programming of heifers employed a 3 × 2 factorial design of maternal (high, H; low, L; and moderate, M) × postnatal (H and L) dietary treatments. Results (Expt. 1) demonstrated that prepubertal heifers born to L dams, regardless of postnatal diet, had reduced expression of the short isoform of ObRc compared to H and M dams, with sporadic effects of undernutrition (L or LL) on ObRb, ObRt, and LRP1. Intravenous administration of oleptin to a selected postpubertal group (HH, MH, LL) of ovariectomized, estradiol-implanted heifers fasted for 56 h (Expt. 2) did not create detectable increases in third ventricle cerebrospinal fluid but increased gonadotropin secretion in all nutritional groups tested. Previous work has shown that leptin enhances gonadotropin secretion during fasting via effects at both hypothalamic and anterior pituitary levels in cattle. Given the apparent lack of robust transfer of leptin across the blood-brain barrier in the current study, effects of leptin at the adenohypophyseal level may predominate in this experimental model.


Asunto(s)
Leptina , Receptores de Leptina , Femenino , Animales , Bovinos , Ovinos , Embarazo , Leptina/genética , Leptina/farmacología , Leptina/metabolismo , Receptores de Leptina/genética , Estado Nutricional , Gonadotropinas/metabolismo , Dieta , Mamíferos/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda