Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Intervalo de año de publicación
1.
Gen Comp Endocrinol ; 336: 114247, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36858273

RESUMEN

The hypothalamic-pituitary-gonadal axis plays a fundamental role in the endocrine regulation of the reproductive function in mammals. Any change in the function of the participating hormones or their receptors can lead to alterations in sexual differentiation, the onset of puberty, infertility, cancer development, and other dysfunctions. In this study, we analyzed the influence of persistently elevated levels of the human chorionic gonadotropin hormone (hCG), a powerful agonist of pituitary luteinizing hormone (LH), on the reproductive axis of female mice. As a consequence of chronic hCG hypersecretion through a global expression of the hCGbeta-subunit in transgenic (TG) female mice, a series of events perturbed the prepubertal to juvenile transition. The imbalance in gonadotropin action was first manifested by precocious puberty and alterations in gonadal hormone production, with the consequent ovarian function disruption and infertility in adulthood. The expansion of cumulus cells in vivo and in vitro, ovulatory capacity, and gene expression of ovulation-related marker genes after hormone stimulation were normal in 3-week-old TG females. However, the expression of genes related to steroidogenesis and luteinization such as Lhcgr, Prlr, and the steroidogenic enzymes Cyp11a1, Cyp17a1, and Cyp19a1 were significantly elevated in the TG females. This study demonstrates that the excessive secretion of hCG in concert with high prolactin, induced premature luteinization, and enhanced ovarian steroidogenesis, as was shown by the up-regulation of luteal cell markers and progesterone synthesis in the TG mice. Furthermore, progressively impaired reproductive function of the TG females occurred from the peripubertal stage to adulthood, thus culminating in infertility.


Asunto(s)
Gonadotropina Coriónica , Infertilidad , Humanos , Ratones , Femenino , Animales , Gonadotropina Coriónica/farmacología , Gonadotropina Coriónica Humana de Subunidad beta/genética , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Ratones Transgénicos , Luteinización , Mamíferos/metabolismo
2.
Am J Physiol Endocrinol Metab ; 324(4): E314-E329, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36652400

RESUMEN

Kisspeptin and γ-amino butyric acid (GABA), synthesized in the central nervous system, are critical for reproduction. Both are also expressed in peripheral organs/tissues critical to metabolic control (liver/pancreas/adipose). Many kisspeptin neurons coexpress GABAB receptors (GABABR) and GABA controls kisspeptin expression and secretion. We developed a unique mouse lacking GABABR exclusively from kisspeptin cells/neurons (Kiss1-GABAB1KO) to evaluate the impact on metabolism/reproduction. We confirmed selective deletion of GABABR from Kiss1 cells in the anteroventral periventricular nucleus/periventricular nucleus continuum (AVPV/PeN; immunofluorescence and PCR) and arcuate nucleus (ARC), medial amygdala (MeA), pituitary, liver, and testes (PCR). Young Kiss1-GABAB1KO males were fertile, with normal LH and testosterone. Kiss1 expression was similar between genotypes in AVPV/PeN, ARC, MeA, bed nucleus of the stria terminalis (BNST), and peripheral organs (testis, liver, pituitary). Kiss1-GABAB1KO males presented higher fasted glycemia and insulin levels, an impaired response to a glucose overload, reduced insulin sensitivity, and marked insulin resistance. Interestingly, when Kiss1-GABAB1KO males got older (9 mo old) their body weight (BW) increased, in part due to an increase in white adipose tissue (WAT). Old Kiss1-GABAB1KO males showed higher fasted insulin, increased pancreatic insulin content, insulin resistance, and significantly decreased pancreatic kisspeptin levels. In sum, lack of GABABR specifically in Kiss1 cells severely impacts glucose homeostasis in male mice, reinforcing kisspeptin involvement in metabolic regulation. These alterations in glucose homeostasis worsened with aging. We highlight the impact of GABA through GABABR in the regulation of the pancreas kisspeptin system in contrast to liver kisspeptin that was not affected.NEW & NOTEWORTHY We developed a unique mouse lacking GABAB receptors specifically in Kiss1 cells to evaluate the impact on reproduction and metabolism. Knockout males showed a severe impact on glucose homeostasis, which worsened with aging. These results reinforce the proposed kisspeptin involvement in metabolic regulation and highlight the impact of GABA through GABABR in the regulation of the peripheral pancreas kisspeptin system.


Asunto(s)
Resistencia a la Insulina , Insulinas , Ratones , Animales , Masculino , Kisspeptinas/genética , Kisspeptinas/metabolismo , Resistencia a la Insulina/genética , Estradiol/metabolismo , Ratones Noqueados , Reproducción/genética , Homeostasis , Ácido gamma-Aminobutírico/metabolismo
3.
Neuroendocrinology ; 112(10): 998-1026, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34963114

RESUMEN

INTRODUCTION: The kisspeptin gene Kiss1 is expressed in two hypothalamic areas: anteroventral periventricular nucleus/periventricular nucleus (AVPV/PeN) and arcuate nucleus (ARC), and also in gonads. Several pieces of evidence suggests that gamma-amino butyric acid B receptors (GABAB) signaling can regulate Kiss1 expression. Here, we inhibited GABAB signaling from PND2 to PND21 and evaluated the hypothalamic-pituitary-gonadal (HPG) axis. METHODS: BALB/c mice were treated on postnatal days 2-21 (PND2-PND21) with CGP55845 (GABAB antagonist) and evaluated in PND21 and adulthood: gene expression (qPCR) in the hypothalamus and gonads, hormones by radioimmunoassay, gonad histochemistry (H&E), puberty onset, and estrous cycles. RESULTS: At PND21, CGP inhibited Kiss1 and Tac2 and increased Pdyn and Gabbr1 in the ARC of both sexes and decreased Th only in female AVPV/PeN. Serum follicle-stimulating hormone (FSH) and testis weight were decreased in CGP-males, and puberty onset was delayed. In adults, Kiss1, Tac2, Pdyn, Pgr, Cyp19a1, and Gad1 were downregulated, while Gabbr1 was upregulated in the ARC of both sexes. In the AVPV/PeN, Kiss1, Th, Cyp19a1, and Pgr were decreased while Gad1 was increased in CGP-females, whereas Cyp19a1 was increased in CGP-males. Serum FSH was increased in CGP-males while prolactin was increased in CGP-females. Testosterone and progesterone were increased in ovaries from CGP-females, in which Kiss1, Cyp19a1, and Esr1 were downregulated while Hsd3b2 was upregulated, together with increased atretic and decreased ovulatory follicles. Testes from CGP-males showed decreased progesterone, increased Gabbr1, Kiss1, Kiss1r, and Esr2 and decreased Cyp19a1, and clear signs of seminiferous tubules atrophy. CONCLUSION: These results demonstrate that appropriate GABAB signaling during this critical prepubertal period is necessary for the normal development of the HPG axis.


Asunto(s)
Kisspeptinas , Progesterona , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Hormona Folículo Estimulante , Antagonistas del GABA , Gónadas , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Ratones , Progesterona/metabolismo , Prolactina/metabolismo , Receptores de Kisspeptina-1/metabolismo , Maduración Sexual/fisiología , Testosterona/metabolismo , Destete
4.
J Comp Physiol B ; 192(1): 141-159, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34459966

RESUMEN

Melatonin, the key messenger of photoperiodic information, is synthesized in the pineal gland by arylalkylamine N-acetyltransferase enzyme (AANAT). It binds to specific receptors MT1 and MT2 located in the hypothalamus and pituitary gland. Melatonin can modulate the reproductive axis affecting the secretion of gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH). The South American plains vizcacha, Lagostomus maximus, shows natural poliovulation of up to 800 oocytes per estrous cycle, a 154-day long pregnancy, and reactivation of the reproductive axis at mid-gestation with pre-ovulatory follicular recruitment, presence of active corpora lutea, and variations of the endocrine status. Here we analyzed the involvement of melatonin in the modulation of the hypothalamic and pituitary gland physiology of vizcacha thorough several approaches, including histological localization of melatoninergic system components, assessment of melatoninergic components expression throughout the reproductive cycle, and evaluation of the effect of melatonin on hypothalamic and pituitary activities during the follicular and luteal phases of the estrous cycle. AANAT and melatonin receptors were localized in the pineal gland and preoptic area of the hypothalamus. Increase in pineal AANAT and serum melatonin expression was observed as pregnancy progressed, with the lowest hypothalamic MT1 and MT2 levels at mid-pregnancy. Pulsatility assays demonstrated that melatonin induces GnRH and LH secretion at luteal phase. The melatoninergic system effects on hypothalamic and pituitary gland hormones secretion during pregnancy pinpoint to melatonin as a potential key factor underlying the reactivation of the reproductive axis activity at mid-gestation.


Asunto(s)
Melatonina , Animales , Femenino , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Melatonina/metabolismo , Hipófisis/metabolismo , Embarazo , América del Sur
5.
Food Chem Toxicol ; 156: 112515, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34400204

RESUMEN

Chlorpyrifos (CPF), the most used insecticide in Argentina, can act as an endocrine disruptor at low doses. We previously demonstrated that chronic exposure to CPF induces hormonal imbalance in vivo. The aim of this work was to study the effects of low concentrations of CPF (0.01 and 1 mg/kg/day) on the reproductive system of virgin adult rats. In the ovary, we studied the effects of CPF on steroidogenesis by determining steroid hormone content by RIA and CYP11 and CYP19 enzyme expression by qRT-PCR. The estrous cycle was evaluated by microscopic observation of vaginal smear, as well as by changes in uterine histology. In endometrium, we determined the fractal dimension and expression of PCNA, ERα and PR by IHC. Our results showed that chronic exposure to CPF affects ovarian steroid synthesis, causing alterations in the normal cyclicity of animals. In addition, CPF induced proliferative changes in the uterus, suggesting that it could affect reproduction or act as a risk factor in the development of uterine proliferative pathologies.


Asunto(s)
Cloropirifos/administración & dosificación , Cloropirifos/toxicidad , Ciclo Estral/efectos de los fármacos , Ovario/efectos de los fármacos , Útero/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Insecticidas/administración & dosificación , Insecticidas/toxicidad , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Vagina/efectos de los fármacos
6.
Reprod Sci ; 28(12): 3547-3561, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33856666

RESUMEN

To explore in mice if a 15% food restriction protocol during pregnancy programs the offspring postnatal development, with emphasis on reproductive function, and to assess if ghrelin (Ghrl) administration to mouse dams exerts effects that mimic those obtained under mild caloric restriction. Mice were 15% food-restricted, injected with 4 nmol/animal/day of Ghrl, or injected with the vehicle (control) thorough pregnancy. After birth, the pups did not receive further treatment. Pups born from food-restricted dams (FR pups) were lighter than Ghrl pups at birth, but reached normal weight at adulthood. Ghrl pups were heavier at birth and gained more weight than control pups (C pups). This effect was not associated with plasma IGF-1. FR pups showed a delay in pinna detachment and eye opening, while an advance was observed in Ghrl pups. FR pups showed also impairment in the surface-righting reflex. In both female FR and Ghrl pups, there was an advance in vaginal opening and, in adulthood, FR pups showed a significant decrease in their own litter size and plasma progesterone, and an increase in embryo loss. A delay in testicular descent was evident in male Ghrl pups. Changes in puberty onset were not associated with differences in the expression of Kiss1 in hypothalamic nuclei. Finally, in adulthood, FR pups showed a significant decrease in sperm quality. In conclusion, a mild food restriction thorough gestation exerted programming effects on the offspring, affecting also their reproductive function in adulthood. These effects were not similar to those of intragestational Ghrl administration.


Asunto(s)
Restricción Calórica/métodos , Desarrollo Fetal/fisiología , Ghrelina/administración & dosificación , Efectos Tardíos de la Exposición Prenatal/genética , Desarrollo Sexual/fisiología , Animales , Animales Recién Nacidos , Vías de Administración de Medicamentos , Femenino , Desarrollo Fetal/efectos de los fármacos , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Desarrollo Sexual/efectos de los fármacos
7.
Mol Neurobiol ; 58(5): 2088-2106, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33411236

RESUMEN

Patients suffering of amyotrophic lateral sclerosis (ALS) present motoneuron degeneration leading to muscle atrophy, dysphagia, and dysarthria. The Wobbler mouse, an animal model of ALS, shows a selective loss of motoneurons, astrocytosis, and microgliosis in the spinal cord. The incidence of ALS is greater in men; however, it increases in women after menopause, suggesting a role of sex steroids in ALS. Testosterone is a complex steroid that exerts its effects directly via androgen (AR) or Sigma-1 receptors and indirectly via estrogen receptors (ER) after aromatization into estradiol. Its reduced-metabolite 5α-dihydrotestosterone acts via AR. This study analyzed the effects of testosterone in male symptomatic Wobblers. Controls or Wobblers received empty or testosterone-filled silastic tubes for 2 months. The cervical spinal cord from testosterone-treated Wobblers showed (1) similar androgen levels to untreated control and (2) increased levels of testosterone, and its 5α-reduced metabolites, 5α- dihydrotestosterone, and 3ß-androstanediol, but (3) undetectable levels of estradiol compared to untreated Wobblers. Testosterone-treated controls showed comparable steroid concentrations to its untreated counterpart. In testosterone- treated Wobblers a reduction of AR, ERα, and aromatase and high levels of Sigma-1 receptor mRNAs was demonstrated. Testosterone treatment increased ChAT immunoreactivity and the antiinflammatory mediator TGFß, while it lessened vacuolated motoneurons, GFAP+ astrogliosis, the density of IBA1+ microgliosis, proinflammatory mediators, and oxidative/nitrosative stress. Clinically, testosterone treatment in Wobblers slowed the progression of paw atrophy and improved rotarod performance. Collectively, our findings indicate an antiinflammatory and protective effect of testosterone in the degenerating spinal cord. These results coincided with a high concentration of androgen-reduced derivatives after testosterone treatment suggesting that the steroid profile may have a beneficial role on disease progression.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Neuronas Motoras/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Testosterona/uso terapéutico , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Aromatasa/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/metabolismo , Masculino , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Fármacos Neuroprotectores/farmacología , Receptores Androgénicos/metabolismo , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/patología , Testosterona/metabolismo , Testosterona/farmacología , Resultado del Tratamiento
8.
Gen Comp Endocrinol ; 296: 113518, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32474048

RESUMEN

In addition to key mammotrophic hormones such as the pituitary prolactin (PRL) and the ovarian steroids progesterone and estradiol, there are local factors that modulate the tissue dynamics of the mammary glands during pregnancy and lactation. By immunohistochemistry and RT-PCR, we found local transcription and translation of gonadotropin-releasing hormone (GNRH), GNRH receptor (GNRHR), PRL and PRL receptor (PRLR) in mammary glands of adult vizcachas during pregnancy and lactation. Both GNRH and GNRHR showed a lag between protein expression and gene transcription throughout the gestational period: while the highest transcription levels of these genes were recorded at early-pregnancy, the epithelial immunoexpressions of both showed their maximum during lactation. RIA results corroborated the presence of GNRH in mammary glands at all the analyzed stages and confirmed the maximum amount of this peptide in the lactating group. Significant amounts of GNRH were detected in milk samples as well. Conversely, PRL and PRLR shared similar protein and gene expression profiles, all exhibiting maximum values during lactation. GNRH peptide content in mammary glands of females with sulpiride-induced hyperprolactinemia (HP) was significantly lower than that of control females (CT). Although PRL mRNA levels remained unchanged, there was a marked increase in theα-lactalbumin (LALBA) transcription in mammary glands of HP- vs CT-females. These results suggest that after targeting mammary glands, PRL stimulates the expression of milk protein genes, but also, tempers the local expression of GNRH. Mammary gland-explantssupplemented with a GNRH analogue (GN-explants) had no differences in terms of PRLR orLALBA transcription levels compared to CT-explants, so the mammary PRLR signaling would not appear to be modulated by GNRH. Yet, mRNA expression levels of both GNRH and the GNRHR-downstream factor, EGR1, were significantly higher in GN-explants compared to that of CT which would point to a GNRH-positive feedback mechanism. In summary, the local coupled expression of GNRH, GNRHR and EGR1 in the mammary gland throughout pregnancy of vizcachas, the PRL-dependent mammary GNRH secretion as well as the GNRH positive feedback on its own transcription suggest an autocrine-paracrine regulatory mechanism and propose an active role for GNRH in mammary gland tissue remodeling.


Asunto(s)
Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/genética , Homeostasis , Glándulas Mamarias Animales/metabolismo , Receptores LHRH/genética , Roedores/genética , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Epitelio/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/metabolismo , Lactancia/fisiología , Ligandos , Especificidad de Órganos , Embarazo , Prolactina/genética , Prolactina/metabolismo , ARN Mensajero/metabolismo , Receptores LHRH/metabolismo , Receptores de Prolactina/genética , Receptores de Prolactina/metabolismo , Reproducción , Transducción de Señal/efectos de los fármacos
9.
Eur J Neurosci ; 52(3): 2995-3001, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32372526

RESUMEN

GnRH neuron activity is under the influence of multiple stimuli, including those coming from the endocannabinoid and the immune systems. Since it has been previously suggested that some of the main elements controlling the GnRH pulse generator possess the TRPV1 receptor, the aim of the present study was to evaluate the participation of the hypothalamic TRPV1, through its pharmacological blockade, in the activity of the hypothalamic-pituitary-testicular axis in male rats under basal or acute inflammatory conditions. Our hypothesis was based on the idea that the hypothalamic TRPV1 participates in the synthesis of the main neuromodulatory signals controlling GnRH, and therefore the reproductive axis. Our results showed that the hypothalamic TRPV1 blockade induced pro-inflammatory effects by increasing Tnfα and Il-1ß mRNA hypothalamic levels and inhibited the reproductive axis by affecting Gnrh, Kiss1 and Rfrp3 mRNA levels and decreasing plasma levels of luteinizing hormone and testosterone under basal conditions, without significant additive effects in rats exposed to systemic LPS. Altogether, these results suggest that the hypothalamic TRPV1 receptor participates in the regulation of the GnRH system, probably by modulating immune-dependent mechanisms.


Asunto(s)
Hormona Liberadora de Gonadotropina , Hormona Luteinizante , Animales , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Masculino , Neuronas/metabolismo , Ratas , Canales Catiónicos TRPV/genética , Testosterona
10.
Am J Physiol Endocrinol Metab ; 318(6): E901-E919, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32286880

RESUMEN

Lack of GABAB receptors in GABAB1 knockout mice decreases neonatal ARC kisspeptin 1 (Kiss1) expression in the arcuate nucleus of the hypothalamus (ARC) in females, which show impaired reproduction as adults. Our aim was to selectively impair GABAB signaling during a short postnatal period to evaluate its impact on the reproductive system. Neonatal male and female mice were injected with the GABAB antagonist CGP 55845 (CGP, 1 mg/kg body wt sc) or saline from postnatal day 2 (PND2) to PND6, three times per day (8 AM, 1 PM, and 6 PM). One group was killed on PND6 for collection of blood samples (hormones by radioimmunoassay), brains for gene expression in the anteroventral periventricular nucleus-periventricular nucleus continuum (AVPV/PeN), and ARC micropunches [quantitative PCR (qPCR)] and gonads for qPCR, hormone contents, and histology. A second group of mice was injected with CGP (1 mg/kg body wt sc) or saline from PND2 to PND6, three times per day (8 AM, 1 PM, and 6 PM), and left to grow to adulthood. We measured body weight during development and parameters of sexual differentiation, puberty onset, and estrous cycles. Adult mice were killed, and trunk blood (hormones), brains for qPCR, and gonads for qPCR and hormone contents were obtained. Our most important findings on PND6 include the CGP-induced decrease in ARC Kiss1 and increase in neurokinin B (Tac2) in both sexes; the decrease in AVPV/PeN tyrosine hydroxylase (Th) only in females; the increase in gonad estradiol content in both sexes; and the increase in primordial follicles and decrease in primary and secondary follicles. Neonatally CGP-treated adults showed decreased ARC Kiss1 and ARC gonadotropin-releasing hormone (Gnrh1) and increased ARC glutamic acid decarboxylase 67 (Gad1) only in males; increased ARC GABAB receptor subunit 1 (Gabbr1) in both sexes; and decreased AVPV/PeN Th only in females. We demonstrate that ARC Kiss1 expression is chronically downregulated in males and that the normal sex difference in AVPV/PeN Th expression is abolished. In conclusion, neonatal GABAergic input through GABAB receptors shapes gene expression of factors critical to reproduction.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipotálamo Anterior/metabolismo , Receptores de GABA-B/metabolismo , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Estradiol/metabolismo , Femenino , Hormona Folículo Estimulante/metabolismo , Antagonistas de Receptores de GABA-B/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo Anterior/efectos de los fármacos , Kisspeptinas/genética , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ovario/efectos de los fármacos , Ovario/metabolismo , Ácidos Fosfínicos/farmacología , Propanolaminas/farmacología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Pubertad/efectos de los fármacos , Pubertad/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores de GABA-B/genética , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Reproducción/efectos de los fármacos , Reproducción/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Diferenciación Sexual/efectos de los fármacos , Diferenciación Sexual/genética , Taquicininas/genética , Taquicininas/metabolismo , Testículo/efectos de los fármacos , Testículo/metabolismo , Testosterona/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
11.
J Steroid Biochem Mol Biol ; 200: 105627, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32070756

RESUMEN

The South American plains vizcacha, Lagostomus maximus, is the only mammal described so far that shows expression of estrogen receptors (ERs) and progesterone receptors (PRs) in gonadotropin-releasing hormone (GnRH) neurons. This animal therefore constitutes an exceptional model for the study of the effect of steroid hormones on the modulation of the hypothalamic-pituitary-ovarian (HPO) axis. By using both in vivo and ex vivo approaches, we have found that pharmacological doses of progesterone (P4) and estradiol (E2) produced an inhibition in the expression of hypothalamic GnRH, while physiological doses produced a differential effect on the pulsatile release frequency or genomic expression of GnRH. Our ex vivo experiment indicates that a short-term effect of E2 modulates the frequency of GnRH release pattern that would be associated with membrane ERs. On the other hand, our in vivo approach suggests that a long-term effect of E2, acting through the classical nuclear ERs-PRs pathway, would produce the modification of GnRH mRNA expression during the GnRH pre-ovulatory surge. Particularly, P4 induced a rise in GnRH mRNA expression and protein release with a decrease in its release frequency. These results suggest different levels of action of steroid hormones on GnRH modulation. We conclude that the fine action of E2 and P4 constitute the key factor to enable the hypothalamic activity during the pregnancy of this mammal.


Asunto(s)
Estradiol/farmacología , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Progesterona/farmacología , Animales , Estradiol/sangre , Femenino , Hormona Liberadora de Gonadotropina/genética , Sistema Hipotálamo-Hipofisario , Hipotálamo/metabolismo , Hormona Luteinizante/metabolismo , Ovariectomía , Ovario , Progesterona/sangre , Roedores
12.
J Neuroendocrinol ; 31(8): e12765, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31269532

RESUMEN

Female mice lacking GABAB receptors, GABAB1KO, show disrupted oestrous cycles, reduced pregnancies and increased hypothalamic Gnrh1 mRNA expression, whereas anteroventral periventricular/periventricular preoptic nucleus (AVPV/PeN) Kiss1 mRNA was not affected. In the present study, we characterise the important components of the gonadotrophic preovulatory surge, aiming to unravel the origin of this reproductive impairment. In GABAB1KO and wild-type (WT) females, we determined: (i) hypothalamic oestrogen receptor (ER)α and ß and aromatase mRNA and protein expression; (ii) ovulation index and oestrus serum follicle-stimulating hormone (FSH) and pituitary Gnrh1r expression; (iii) in ovariectomised-oestradiol valerate-treated mice, we evaluated ex vivo hypothalamic gonadotrophin-releasing hormone (GnRH) pulsatility in the presence/absence of kisspeptin (Kiss-10, constant or pulsatile) and oestradiol (constant); and (iv) in ovariectomised-oestradiol silastic capsule-treated mice (proestrous-like environment), we evaluated morning and evening kisspeptin neurone activation (c-Fos+) and serum luteinising homrone (LH). In the medial basal hypothalamus of oestrus GABAB1KOs, aromatase and ERα mRNA and protein were increased, whereas ERß was decreased. In GABAB1KOs, the ovulation index was decreased together with decreased first oestrus serum FSH and increased pituitary Gnrh1r mRNA. Under constant Kiss-10 stimulation, hypothalamic GnRH pulse frequency did not vary, although GnRH mass/pulse was increased in GABAB1KOs. In WTs, pulsatile Kiss-10 together with constant oestradiol significantly increased GnRH pulsatility, whereas, in GABAB1KOs, oestradiol alone increased GnRH pulsatility and this was reversed by pulsatile Kiss-10 addition. In GABAB1KOs AVPV/PeN kisspeptin neurones were similarly activated (c-Fos+) in the morning and evening, whereas WTs showed the expected, marked evening stimulation. LH correlated with activated kisspeptin cells in WT mice, whereas GABAB1KO mice showed high, similar LH levels both in the morning and evening. Taken together, all of these alterations point to impairment in the trigger of the preovulatory GnRH surge that entails the reproductive alterations described.


Asunto(s)
Ciclo Estral/sangre , Ciclo Estral/genética , Hormona Luteinizante/sangre , Inhibición de la Ovulación , Receptores de GABA-B/genética , Animales , Estradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Hormona Liberadora de Gonadotropina/sangre , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ovulación/sangre , Ovulación/genética , Inhibición de la Ovulación/sangre , Inhibición de la Ovulación/genética , Regulación hacia Arriba/genética
13.
Reproduction ; 157(6): R225-R233, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30844750

RESUMEN

Neuroendocrine control of reproduction involves the interplay of various factors that become active at some point along development. GnRH is the main neurohormone controlling reproduction and among the most important inputs modulating GnRH synthesis/secretion are GABA and kisspeptins. These interactions of GABA and kisspeptin in the control of GnRH secretion can take place by the presence of the receptors of both factors on the GnRH neuron or alternatively by the actions of GABA on kisspeptin neurons and/or the actions of kisspeptin on GABA neurons. Kisspeptin acts on the Kiss1R, a seven transmembrane domain, Gαq/11-coupled receptor that activates phospholipase C, although some Gαq/11-independent pathways in mediating part of the effects of Kiss1R activation have also been proposed. GABA acts through two kinds of receptors, ionotropic GABAA/C receptors involving a chloride channel and associated with fast inhibitory/stimulatory conductance and metabotropic GABAB receptors (GABABR) that are Gi/0 protein linked inducing late slow hyperpolarization. In this review, we aim to summarize the different ways in which these two actors, kisspeptin and GABA, interact to modulate GnRH secretion across the reproductive lifespan.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Receptores de Kisspeptina-1/metabolismo , Reproducción/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Humanos
14.
Gen Comp Endocrinol ; 273: 40-51, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29656043

RESUMEN

The South American plains vizcacha, Lagostomus maximus, is a caviomorph rodent native from Argentina, Bolivia and Paraguay. It shows peculiar reproductive features like pre-ovulatory follicle recruitment during pregnancy with an ovulatory process at around mid-gestation. We have described the activation of the hypothalamic - pituitary - ovarian (HPO) axis during pregnancy. A progressive decrease of progesterone (P4) at mid-pregnancy elicits the delivery of gonadotropin-releasing hormone (GnRH) with the consequent secretion of follicle stimulating hormone (FSH) and estradiol (E2) followed by luteinizing hormone (LH) release resulting in follicular luteinization and the P4 concentration recover. Pituitary gland is the central regulator of the HPO axis being E2 a key hormone involved in the regulation of its activity. In this work we analyzed the action of E2 on the pituitary response to the GnRH wave as well as its involvement on LH secretion at mid-gestation in L. maximus. The expression of GnRHR at the pituitary pars distalis showed a significant decrease at mid-pregnancy compared to early- and term-gestating females. ERα showed a significant increment from mid-gestation whereas ERß did not show variations throughout pregnancy; whereas the LH expression in the pituitary pars distalis showed a significant increase at mid-gestation, concordantly with serum LH, which was followed by a decrease at term-gestation with similar values than at early-pregnancy. The number of cells with co-localization of ERα and GnRHR showed a decline at mid-pregnancy related to early- and term-gestation, whereas the cells with co-localization of ERα and LH increased at mid- and term-pregnancy. On the other hand, ex vivo measuring of LH pulsatility showed a significant increment in the total mass of LH delivered at mid-pregnancy followed by a decrease at term-gestation. The stimulation of ERα with the PPT specific agonist induced a significant increment in the total mass of LH released, whereas no changes were determined when ERß was stimulated with its specific agonist MPP. These results suggest that LH pulsatility rise at mid-pregnancy would be enabled by the increase of E2 acting through ERα.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Hormona Luteinizante/metabolismo , Hipófisis/metabolismo , Roedores/metabolismo , Animales , Antineoplásicos Hormonales , Receptor beta de Estrógeno/metabolismo , Femenino , Adenohipófisis/metabolismo , Embarazo , Receptores LHRH/metabolismo
15.
Endocrinology ; 159(9): 3389-3402, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30107405

RESUMEN

Kisspeptin, encoded by Kiss1, activates reproduction by stimulating GnRH neurons. Although most Kiss1 neurons are located in the hypothalamus, smaller Kiss1 populations also reside in the medial amygdala (MeA), bed nucleus of the stria terminalis (BnST), and lateral septum (LS). However, very little is known about the regulation and function of these extra-hypothalamic Kiss1 neurons. This study focused on the roles and interactions of two signaling factors, estradiol (E2) and GABA, known to stimulate and inhibit, respectively, extra-hypothalamic Kiss1 expression. First, using estrogen receptor (ER)α knockout (KO) and ßERKO mice, we demonstrated that Kiss1 in both the BnST and LS is stimulated by E2, as occurs in the MeA, and that this E2 upregulation occurs via ERα, but not ERß. Second, using GABABR KO and wild-type mice, we determined that whereas E2 normally increases extra-hypothalamic Kiss1 levels, such upregulation by E2 is further enhanced by the concurrent absence of GABABR signaling in the MeA and LS, but not the BnST. Third, we demonstrated that when GABABR signaling is absent, the additional removal of gonadal sex steroids does not abolish Kiss1 expression in the MeA and BnST, and in some cases the LS. Thus, Kiss1 expression in these extra-hypothalamic regions is not solely dependent on E2 stimulation. Finally, we demonstrated a significant positive correlation between Kiss1 levels in the MeA, BnST, and LS, but not between these regions and the hypothalamus (anteroventral periventricular nucleus/periventricular nucleus). Collectively, our findings indicate that both E2 and GABA independently regulate all three extra-hypothalamic Kiss1 populations, but their regulatory interactions may vary by brain region and additional yet-to-be-identified factors are likely involved.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Estradiol/farmacología , Estrógenos/farmacología , Kisspeptinas/genética , Neuronas/efectos de los fármacos , Receptores de GABA-B/metabolismo , Núcleos Septales/efectos de los fármacos , Amígdala del Cerebelo/citología , Amígdala del Cerebelo/metabolismo , Animales , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Kisspeptinas/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Núcleos Septales/citología , Núcleos Septales/metabolismo , Transducción de Señal , Ácido gamma-Aminobutírico/metabolismo
16.
Physiol Rep ; 5(19)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29038356

RESUMEN

Females of the South American plains vizcacha, Lagostomus maximus, show peculiar reproductive features such as massive polyovulation up to 800 oocytes per estrous cycle and an ovulatory process around mid-gestation arising from the reactivation of the hypothalamic-hypophyseal-ovary (H.H.O.) axis. Estradiol (E2) regulates gonadotropin-releasing hormone (GnRH) expression. Biosynthesis of estrogens results from the aromatization of androgens by aromatase, which mainly occurs in the gonads, but has also been described in the hypothalamus. The recently described correlation between GnRH and ERα expression patterns in the hypothalamus of the vizcacha during pregnancy, with coexpression in the same neurons of the medial preoptic area, suggests that hypothalamic synthesis of E2 may affect GnRH neurons and contribute with systemic E2 to modulate GnRH delivery during the gestation. To elucidate this hypothesis, hypothalamic expression and the action of aromatase on GnRH release were evaluated in female vizcachas throughout pregnancy. Aromatase and GnRH expression was increased significantly in mid-pregnant and term-pregnant vizcachas compared to early-pregnant and nonpregnant females. In addition, aromatase and GnRH were colocalized in neurons of the medial preoptic area of the hypothalamus throughout gestation. The blockage of the negative feedback of E2 induced by the inhibition of aromatase resulted in a significant increment of GnRH-secreted mass by hypothalamic explants. E2 produced in the same neurons as GnRH may drive intracellular E2 to higher levels than those obtained from systemic circulation alone. This may trigger for a prompt GnRH availability enabling H.H.O. activity at mid-gestation with ovulation and formation of accessory corpora lutea with steroidogenic activity that produce the necessary progesterone to maintain gestation to term and guarantee the reproductive success.


Asunto(s)
Estradiol/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/metabolismo , Embarazo/metabolismo , Animales , Aromatasa/metabolismo , Retroalimentación Fisiológica , Femenino , Hipotálamo/citología , Neuronas/metabolismo , Roedores
17.
J Mol Histol ; 48(3): 259-273, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28317066

RESUMEN

Gonadotropin-releasing hormone (GnRH) is the key regulator of the hypothalamic-pituitary-gonadal axis. Estradiol (E2) affects GnRH synthesis and delivery. Hypothalamic estrogen receptors (ER) modulate GnRH expression acting as transcription factors. The South American plains vizcacha, Lagostomus maximus, is able to ovulate up to 800 oocytes per reproductive cycle, and shows continuous folliculogenesis with pre-ovulatory follicle formation and an ovulatory event at mid-gestation. The aim of this work was to analyze the hypothalamic expression of ER in the vizcacha at different gestational time-points, and its relationship with GnRH expression, serum luteinizing hormone (LH) and E2. The hormonal pattern of mid-gestating vizcachas was comparable to ovulating-females with significant increases in GnRH, LH and E2. Hypothalamic protein and mRNA expression of ERα varied during pregnancy with a significant increase at mid-gestation whereas ERß mRNA expression did not show significant variations. Hypothalamic immunolocalization of ERα was observed in neurons of the diagonal band of Brocca, medial preoptic area (mPOA), periventricular, suprachiasmatic, supraoptic (SON), ventromedial, and arcuate nuclei, and medial eminence, with a similar distribution throughout gestation. In addition, all GnRH neurons of the mPOA and SON showed ERα expression with no differences across the reproductive status. The correlation between GnRH and ERα at mid-gestation, and their co-localization in the hypothalamic neurons of the vizcacha, provides novel information compared with other mammals suggesting a direct action of estrogen as part of a differential reproductive strategy to assure GnRH synthesis during pregnancy.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/citología , Neuronas/química , Animales , Estradiol/metabolismo , Femenino , Edad Gestacional , Hormona Luteinizante/sangre , Embarazo , Roedores
18.
Endocrinology ; 157(11): 4446-4460, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27571131

RESUMEN

The Wobbler mouse is an animal model for human motoneuron diseases, especially amyotrophic lateral sclerosis (ALS), used in the investigation of both pathology and therapeutic treatment. ALS is a fatal neurodegenerative disease, characterized by the selective and progressive death of motoneurons, leading to progressive paralysis. Previous limited studies have reported steroidal hormone dysregulation in Wobbler mouse and in ALS patients, suggesting endocrine dysfunctions which may be involved in the pathogenesis of the disease. In this study, we established a steroid profiling in brain, spinal cord, plasma, adrenal glands, and testes in 2-month-old male Wobbler mice and their littermates by gas chromatography coupled to mass spectrometry. Our results show in Wobbler mice the following: 1) a marked up-regulation of corticosterone levels in adrenal glands, plasma, spinal cord regions (cervical, thoracic, lumbar) and brain; 2) a strong decrease in T levels in the testis, plasma, spinal cord, and brain; and 3) increased levels of progesterone and especially of its reduced metabolites 5α-dihydroprogesterone, allopregnanolone, and 20α-dihydroprogesterone in the brain, spinal cord, and adrenal glands. Furthermore, Wobbler mice showed a hypothalamic-pituitary-gonadal hypoactivity. Interestingly, plasma concentrations of corticosterone and T correlate well with their respective levels in cervical spinal cord in both control and Wobbler mice. T down-regulation is probably the consequence of adrenal hyperactivity, and the up-regulation of progesterone and its reduced metabolites may correspond to an endogenous protective mechanism in response to motoneuron degeneration. Our findings suggest that increased levels of corticosterone and decreased levels of T in plasma could be a signature of motoneuron degeneration.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , 17-Cetosteroides/sangre , 17-Cetosteroides/metabolismo , Glándulas Suprarrenales/metabolismo , Esclerosis Amiotrófica Lateral/sangre , Androstanoles/sangre , Androstanoles/metabolismo , Animales , Encéfalo/metabolismo , Corticosterona/sangre , Corticosterona/metabolismo , Modelos Animales de Enfermedad , Femenino , Cromatografía de Gases y Espectrometría de Masas , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante/sangre , Masculino , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/fisiología , Pregnanolona/sangre , Pregnanolona/metabolismo , Progesterona/sangre , Progesterona/metabolismo , Médula Espinal/metabolismo , Testículo/metabolismo , Testosterona/sangre , Testosterona/metabolismo
19.
Gen Comp Endocrinol ; 232: 174-84, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26704854

RESUMEN

Gonadotropin-releasing hormone (GnRH) is the regulator of the hypothalamic-hypophyseal-gonadal (HHG) axis. GnRH and GAP (GnRH-associated protein) are both encoded by a single preprohormone. Different variants of GnRH have been described. In most mammals, GnRH is secreted in a pulsatile manner that stimulates the release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH). The South-American plains vizcacha, Lagostomus maximus, is a rodent with peculiar reproductive features including natural poly-ovulation up to 800 oocytes per estrous cycle, pre-ovulatory follicle formation throughout pregnancy and an ovulatory process which takes place at mid-gestation and adds a considerable number of secondary corpora lutea. Such features should occur under a special modulation of the HHG axis, guided by GnRH. The aim of this study was to sequence hypothalamic GnRH preprogonadotrophin mRNA in the vizcacha, to compare it with evolutionarily related species and to identify its expression, distribution and pulsatile pattern of secretion. The GnRH1variant was detected and showed the highest homology with that of chinchilla, its closest evolutionarily related species. Two isoforms of transcripts were identified, carrying the same coding sequence, but different 5' untranslated regions. This suggests a sensitive equilibrium between RNA stability and translational efficiency. A predominant hypothalamic localization and a pulsatile secretion pattern of one pulse of GnRH every hour were found. The lower homology found for GAP, also among evolutionarily related species, depicts a potentially different bioactivity.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Animales , Femenino , Embarazo , Análisis de Secuencia , América del Sur , Distribución Tisular
20.
Mol Cell Endocrinol ; 390(1-2): 102-11, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24755422

RESUMEN

Transgenic female mice overexpressing the α- and ß- subunits of human chorionic gonadotropin (hCGαß+) exhibited precocious puberty, as evidenced by early vaginal opening. Chronically elevated hCG in 21-day-old hCGαß+ females stimulated gonadal androgen production, which exerted negative feedback over the endogenous gonadotropin synthesis, and activated the hypothalamic GnRH pulsatility and gene expression. Transgenic females also exhibited elevated hypothalamic aromatization in the preoptic area (POA), which is the sexually-differentiated area that controls the LH surge in adulthood. Ovariectomy at 14 days of age was unable to rescue this phenotype. However, the blockade of androgen action by flutamide from postnatal day 6 onwards reduced the aromatase levels in the POA of hCGαß+ females. Our results suggest that early exposure of females to androgen action during a critical period between postnatal days 6-14 induces sex-specific organizational changes of the brain, which affect the aromatase expression in the POA at the onset of precocious puberty.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Hipotálamo/metabolismo , Pubertad Precoz/metabolismo , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Animales , Aromatasa/metabolismo , Células Cultivadas , Gonadotropina Coriónica/fisiología , Estradiol/sangre , Femenino , Flutamida/farmacología , Flutamida/uso terapéutico , Hormona Folículo Estimulante/sangre , Expresión Génica , Hormona Liberadora de Gonadotropina/fisiología , Humanos , Ratones Transgénicos , Hipófisis/metabolismo , Pubertad Precoz/tratamiento farmacológico , Testosterona/sangre , Vagina/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA