Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Anim Reprod Sci ; 266: 107516, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38823233

RESUMEN

Irisin is a hormone secreted by muscle in response to exercise. The irisin receptor (IrisinR) is a heterodimer of integrin alpha V (ITGAV) and integrin beta 5 (ITGB5) subunits. Since irisin may mediate some beneficial effects of exercise on animal reproduction, we tested the hypothesis that bovine gonadotrophs express IrisinR and irisin stimulates luteinizing hormone (LH) and follicle stimulating hormone (FSH) secretion by gonadotrophs. Reverse transcription polymerase chain reaction was used to detect the mRNA expression of both ITGAV and ITGB5 in the anterior pituitary glands (APs) of post pubertal heifers and mouse gonadotroph cell line "LßT2." Western blotting was used to detect protein expression in bovine APs. Immunofluorescence microscopy, utilizing the same antibody, visualized IrisinR on the plasma membrane of majority of gonadotrophs. We prepared AP cells from healthy postpubertal heifers, cultured them for 3.5 d, and treated them with increasing concentrations (0, 0.01, 0.1, 1, or 10 nM) of irisin for 5 min before either no treatment or gonadotropin-releasing hormone (GnRH) stimulation. After 2 h, media were harvested for LH and FSH assays. Irisin (0.1-10 nM) stimulated basal LH and FSH secretion, and these stimulatory effects were inhibited by the extracellular signal-regulated kinase or SMAD pathway inhibitors. In the presence of GnRH, irisin at 0.01-1 nM stimulated LH and FSH secretion. A higher dose of irisin (10 nM), however, suppressed the GnRH-induced LH and FSH levels. In conclusion, bovine gonadotrophs expressed IrisinR, and irisin controlled LH and FSH secretion from bovine gonadotrophs.


Asunto(s)
Fibronectinas , Hormona Folículo Estimulante , Gonadotrofos , Hormona Luteinizante , Animales , Bovinos , Femenino , Ratones , Fibronectinas/metabolismo , Hormona Folículo Estimulante/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Gonadotrofos/metabolismo , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante/metabolismo , Condicionamiento Físico Animal/fisiología
2.
Exp Mol Med ; 53(8): 1238-1249, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34453106

RESUMEN

P62 is a protein adaptor for various metabolic processes. Mice that lack p62 develop adult-onset obesity. However, investigations on p62 in reproductive dysfunction are rare. In the present study, we explored the effect of p62 on the reproductive system. P62 deficiency-induced reproductive dysfunction occurred at a young age (8 week old). Young systemic p62 knockout (p62-/-) and pituitary-specific p62 knockout (p62flox/flox αGSUcre) mice both presented a normal metabolic state, whereas they displayed infertility phenotypes (attenuated breeding success rates, impaired folliculogenesis and ovulation, etc.) with decreased luteinizing hormone (LH) expression and production. Consistently, in an infertility model of polycystic ovary syndrome (PCOS), pituitary p62 mRNA was positively correlated with LH levels. Mechanistically, p62-/- pituitary RNA sequencing showed a significant downregulation of the mitochondrial oxidative phosphorylation (OXPHOS) pathway. In vitro experiments using the pituitary gonadotroph cell line LßT2 and siRNA/shRNA/plasmid confirmed that p62 modulated LH synthesis and secretion via mitochondrial OXPHOS function, especially Ndufa2, a component molecule of mitochondrial complex I, as verified by Seahorse and rescue tests. After screening OXPHOS markers, Ndufa2 was found to positively regulate LH production in LßT2 cells. Furthermore, the gonadotropin-releasing hormone (GnRH)-stimulating test in p62flox/flox αGSUcre mice and LßT2 cells illustrated that p62 is a modulator of the GnRH-LH axis, which is dependent on intracellular calcium and ATP. These findings demonstrated that p62 deficiency in the pituitary impaired LH production via mitochondrial OXPHOS signaling and led to female infertility, thus providing the GnRH-p62-OXPHOS(Ndufa2)-Ca2+/ATP-LH pathway in gonadotropic cells as a new theoretical basis for investigating female reproductive dysfunction.


Asunto(s)
Infertilidad Femenina/metabolismo , Infertilidad Femenina/patología , Hormona Luteinizante/biosíntesis , Proteína Sequestosoma-1/deficiencia , Envejecimiento/patología , Animales , Regulación hacia Abajo/efectos de los fármacos , Femenino , Gonadotrofos/efectos de los fármacos , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Infertilidad Femenina/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Fosforilación Oxidativa/efectos de los fármacos , Reproducción/efectos de los fármacos , Proteína Sequestosoma-1/metabolismo , Transducción de Señal/efectos de los fármacos
3.
Endocr J ; 68(9): 1091-1100, 2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-33994401

RESUMEN

Anti-Müllerian hormone (AMH) is primarily produced by ovarian granulosa cells and contributes to follicle development. AMH is also produced in other tissues, including the brain and pituitary; however, its roles in these tissues are not well understood. In this study, we examined the effect of AMH on pituitary gonadotrophs. We detected AMH and AMH receptor type 2 expression in LßT2 cells. In these cells, the expression of FSHß- but not α- and LHß-subunits increased significantly as the concentration of AMH increased. LßT2 cells expressed Kiss-1 and Kiss-1R. AMH stimulation resulted in decreases in both Kiss-1 and Kiss-1R. The siRNA-mediated knockdown of Kiss-1 in LßT2 cells did not alter the basal expression levels of α-, LHß-, and FSHß-subunits. In LßT2 cells overexpressing Kiss-1R, exogenous kisspeptin stimulation significantly increased the expression of all three gonadotropin subunits. However, kisspeptin-induced increases in these subunits were almost completely eliminated in the presence of AMH. In contrast, GnRH-induced increases in the three gonadotropin subunits were not modulated by AMH. Our observations suggested that AMH acts on pituitary gonadotrophs and induces FSHß-subunit expression with concomitant decreases in Kiss-1 and Kiss-1R gene expression. Kisspeptin, but not GnRH-induced gonadotropin subunit expression, was inhibited by AMH, suggesting that it functions in association with the kisspeptin/Kiss-1R system in gonadotrophs.


Asunto(s)
Hormona Antimülleriana/farmacología , Gonadotrofos/metabolismo , Gonadotropinas Hipofisarias/genética , Kisspeptinas/fisiología , Receptores de Kisspeptina-1/fisiología , Animales , Línea Celular , Hormona Folículo Estimulante de Subunidad beta/genética , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Kisspeptinas/genética , Hormona Luteinizante de Subunidad beta/genética , Ratones , ARN Interferente Pequeño , Receptores de Kisspeptina-1/genética
4.
Mol Cell Endocrinol ; 530: 111286, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-33872733

RESUMEN

Within pituitary gonadotropes, the gonadotropin-releasing hormone receptor (GnRHR) receives hypothalamic input from GnRH neurons that is critical for reproduction. Previous studies have suggested that androgens may regulate GnRHR, although the mechanisms remain unknown. In this study, we demonstrated that androgens positively regulate Gnrhr mRNA in mice. We then investigated the effects of androgens and androgen receptor (AR) on Gnrhr promoter activity in immortalized mouse LßT2 cells, which represent mature gonadotropes. We found that AR positively regulates the Gnrhr proximal promoter, and that this effect requires a hormone response element (HRE) half site at -159/-153 relative to the transcription start site. We also identified nonconsensus, full-length HREs at -499/-484 and -159/-144, which are both positively regulated by androgens on a heterologous promoter. Furthermore, AR associates with the Gnrhr promoter in ChIP. Altogether, we report that GnRHR is positively regulated by androgens through recruitment of AR to the Gnrhr proximal promoter.


Asunto(s)
Andrógenos/farmacología , Gonadotrofos/citología , Receptores Androgénicos/metabolismo , Receptores LHRH/genética , Animales , Línea Celular , Secuenciación de Inmunoprecipitación de Cromatina , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Gonadotrofos/efectos de los fármacos , Gonadotrofos/metabolismo , Masculino , Ratones , Regiones Promotoras Genéticas , Receptores LHRH/metabolismo , Análisis de Secuencia de ADN
5.
Sci Rep ; 11(1): 4757, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637828

RESUMEN

Ethanolamine plasmalogens (EPls), unique alkenylacyl-glycerophospholipids, are the only known ligands of G-protein-coupled receptor 61-a novel receptor co-localised with gonadotropin-releasing hormone receptors on anterior pituitary gonadotrophs. Brain EPl decreases with age. Commercial EPl-extracted from the cattle brain (unidentified age)-can independently stimulate FSH secretion from gonadotrophs. We hypothesised that there exists an age-related difference in the quality, quantity, and ability of bovine brain EPls to stimulate bovine gonadotrophs. We compared the brains of young (about 26 month old heifers) and old (about 90 month old cows) Japanese Black bovines, including EPls obtained from both groups. Additionally, mRNA expressions of the EPl biosynthesis enzymes, glyceronephosphate O-acyltransferase, alkylglycerone phosphate synthase, and fatty acyl-CoA reductase 1 (FAR1) were evaluated in young and old hypothalami. The old-brain EPl did not stimulate FSH secretion from gonadotrophs, unlike the young-brain EPl. Molecular species of EPl were compared using two-dimensional liquid chromatography-mass spectrometry. We identified 20 EPl molecular species of which three and three exhibited lower (P < 0.05) and higher (P < 0.05) ratios, respectively, in old compared to young brains. In addition, quantitative reverse transcription-polymerase chain reaction detected higher FAR1 levels in the POA, but not in the ARC&ME tissues, of old cows than that of fertile young heifers. Therefore, old-brain EPl may be associated with age-related infertility.


Asunto(s)
Factores de Edad , Gonadotrofos/efectos de los fármacos , Plasmalógenos/metabolismo , Plasmalógenos/farmacología , Animales , Encéfalo/metabolismo , Bovinos , Femenino , Hormona Folículo Estimulante/metabolismo , Regulación de la Expresión Génica , Hipotálamo/química , Hipotálamo/enzimología , Plasmalógenos/química
6.
Front Endocrinol (Lausanne) ; 11: 605068, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33365013

RESUMEN

The pituitary gland controls many important physiological processes in vertebrates, including growth, homeostasis, and reproduction. As in mammals, the teleost pituitary exhibits a high degree of plasticity. This plasticity permits changes in hormone production and secretion necessary to meet the fluctuating demands over the life of an animal. Pituitary plasticity is achieved at both cellular and population levels. At the cellular level, hormone synthesis and release can be regulated via changes in cell composition to modulate both sensitivity and response to different signals. At the cell population level, the number of cells producing a given hormone can change due to proliferation, differentiation of progenitor cells, or transdifferentiation of specific cell types. Gonadotropes, which play an important role in the control of reproduction, have been intensively investigated during the last decades and found to display plasticity. To ensure appropriate endocrine function, gonadotropes rely on external and internal signals integrated at the brain level or by the gonadotropes themselves. One important group of internal signals is the sex steroids, produced mainly by the gonadal steroidogenic cells. Sex steroids have been shown to exert complex effects on the teleost pituitary, with differential effects depending on the species investigated, physiological status or sex of the animal, and dose or method of administration. This review summarizes current knowledge of the effects of sex steroids (androgens and estrogens) on gonadotrope cell plasticity in teleost anterior pituitary, discriminating direct from indirect effects.


Asunto(s)
Plasticidad de la Célula , Hormonas Esteroides Gonadales/farmacología , Gonadotrofos/efectos de los fármacos , Hipófisis/efectos de los fármacos , Animales , Peces
7.
Microsc Microanal ; 26(4): 699-707, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32519626

RESUMEN

The aim of the study was to investigate the effects of chronic nandrolone decanoate treatment and/or swimming training on immunohistomorphometric parameters on rat pituitary gonadotropic cells. Male Wistar albino rats, 10 weeks old, were classified into four groups: control (T−N−), nandrolone (T−N+), swimming training (T+N−), and swimming training with nandrolone (T+N+). The T+ groups swam for 4 weeks, 1 h/day, 5 days/week. The N+ groups received nandrolone decanoate (20 mg/kg) once per week for 4 weeks. Pituitary tissue sections were processed and stained for immunohistochemical analysis and immunofluorescence. The volume density of luteinizing hormone (LH) cells was decreased by 48% in T−N+ and for 35% in the T+N+ group. The volume density of follicle-stimulating hormone (FSH) cells was decreased by 39% in T−N+ and for 30% in T+N+ compared to the control. Nandrolone alone, or combined with swimming training, decreased the number of LH/FSH cells compared to the control. The levels of the immunofluorescent signal of LH/FSH cells were increased in all experimental groups. Nandrolone alone decreased the serum level of LH by 17%, whereas swimming training alone increased FSH levels by 11% compared to the control. Serum levels of testosterone were increased in all experimental groups. Nandrolone alone, or combined with swimming training, decreased immunohistomorphometric parameters of gonadotropic cells, whereas the levels of immunofluorescent signal were increased.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Gonadotrofos/metabolismo , Hormona Luteinizante/metabolismo , Nandrolona Decanoato/farmacología , Congéneres de la Testosterona/farmacología , Animales , Doping en los Deportes/métodos , Técnica del Anticuerpo Fluorescente , Hormona Folículo Estimulante/sangre , Gonadotrofos/citología , Gonadotrofos/efectos de los fármacos , Inmunohistoquímica , Hormona Luteinizante/sangre , Masculino , Ratas , Ratas Wistar , Natación
8.
Mol Cell Endocrinol ; 512: 110851, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32439415

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is thought to play a role in the development and regulation of gonadotrophs. PACAP levels are very high in the rodent fetal pituitary, and decline substantially and rapidly at birth, followed by a significant rise in FSHß and GnRH-R expression. Because there is evidence that PACAP stimulates its own transcription, we propose that this self-regulation is interrupted around the time of birth. To begin to examine the mechanisms for PACAP self-regulation, we used two well-established gonadotroph cell lines, αT3-1 cells and the more mature LßT2 cells which were transfected with a PACAP promoter-reporter construct As in vivo, the basal PACAP transcription level is significantly lower in the more mature LßT2 cells in which basal cAMP signaling is also much reduced. The PACAP promoter was stimulated by PACAP in both cell lines. Treatment with inhibitors of second messenger pathways implicated PKA, PKC and MAPK in PACAP transcription. Three regions of the PACAP promoter were found to confer inhibition or stimulation of PACAP transcription. By inhibiting cAMP response element binding (CREB) activity and mutating a proximal CREB binding site, we found that CREB is essential for promoter activation. Finally, overexpression of PACAP receptor HOP1 isoform, to increase the level in LßT2 cells to that of αT3-1 cells and simulate the E19 pituitary, increased PACAP- stimulated sensitivity and significantly altered downstream gene transcription. These results provide novel insight into the feed-forward regulation of PACAP expression that may help initiate gonadotroph function at birth.


Asunto(s)
Gonadotrofos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Animales , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/genética , Células Cultivadas , Embrión de Mamíferos , Femenino , Gonadotrofos/efectos de los fármacos , Gonadotrofos/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/genética , Ratones , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Embarazo , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
9.
J Reprod Dev ; 66(3): 249-254, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32115468

RESUMEN

GPR120 is a long-chain fatty acid (LCFA) receptor that is specifically expressed in gonadotropes in the anterior pituitary gland in mice. The aim of this study was to investigate whether GPR120 is activated by free fatty acids in the pituitary of mice and mouse immortalized gonadotrope LßT2 cells. First, the effects of palmitate on GPR120, gonadotropic hormone b-subunits, and GnRH-receptor expression in gonadotropes were investigated in vitro. We observed palmitate-induced an increase in Gpr120 mRNA expression and a decrease in follicle-stimulating hormone b-subunit (Fshb) expression in LßT2 cells. Furthermore, palmitate exposure caused the phosphorylation of ERK1/2 in LßT2 cells, but no significant changes were observed in the expression levels of luteinizing hormone b-subunit (Lhb) and gonadotropin releasing hormone-receptor (Gnrh-r) mRNA and number of GPR120 immunoreactive cells. Next, diurnal variation in Gpr120 mRNA expression in the male mouse pituitary gland was investigated using ad libitum and night-time restricted feeding (active phase from 1900 to 0700 h) treatments. In ad libitum feeding group mice, Gpr120 mRNA expression at 1700 h was transiently higher than that measured at other times, and the peak blood non-esterified fatty acid (NEFA) levels were observed from 1300 to 1500 h. These results were not observed in night-time-restricted feeding group mice. These results suggest that GPR120 is activated by LCFAs to regulate follicle stimulating hormone (FSH) synthesis in the mouse gonadotropes.


Asunto(s)
Ácidos Grasos no Esterificados/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Gonadotrofos/metabolismo , Ácido Palmítico/farmacología , Hipófisis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Línea Celular , Hormona Folículo Estimulante de Subunidad beta/genética , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Gonadotrofos/efectos de los fármacos , Hormona Luteinizante de Subunidad beta/genética , Hormona Luteinizante de Subunidad beta/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Fosforilación/efectos de los fármacos , Hipófisis/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores LHRH/genética , Receptores LHRH/metabolismo
10.
J Endocrinol ; 245(1): 21-37, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31977313

RESUMEN

Follicle-stimulating hormone (Fsh) and luteinizing hormone (Lh) produced by the gonadotropes play a major role in control of reproduction. Contrary to mammals and birds, Lh and Fsh are mostly produced by two separate cell types in teleost. Here, we investigated gonadotrope plasticity, using transgenic lines of medaka (Oryzias latipes) where DsRed2 and hrGfpII are under the control of the fshb and lhb promotors respectively. We found that Fsh cells appear in the pituitary at 8 dpf, while Lh cells were previously shown to appear at 14 dpf. Similar to Lh cells, Fsh cells show hyperplasia from juvenile to adult stages. Hyperplasia is stimulated by estradiol. Both Fsh and Lh cells show hypertrophy during puberty with similar morphology. They also share similar behavior, using their cellular extensions to make networks. We observed bi-hormonal gonadotropes in juveniles and adults but not in larvae where only mono-hormonal cells are observed, suggesting the existence of phenotypic conversion between Fsh and Lh in later stages. This is demonstrated in cell culture, where some Fsh cells start to produce Lhß, a phenomenon enhanced by gonadotropin-releasing hormone (Gnrh) stimulation. We have previously shown that medaka Fsh cells lack Gnrh receptors, but here we show that with time in culture, some Fsh cells start responding to Gnrh, while fshb mRNA levels are significantly reduced, both suggestive of phenotypic change. All together, these results reveal high plasticity of gonadotropes due to both estradiol-sensitive proliferation and Gnrh promoted phenotypic conversion, and moreover, show that gonadotropes lose part of their identity when kept in cell culture.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Gonadotrofos/metabolismo , Hormona Luteinizante/metabolismo , Oryzias/metabolismo , Maduración Sexual/fisiología , Animales , Animales Modificados Genéticamente , Células Cultivadas , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Hormona Folículo Estimulante/genética , Expresión Génica , Gonadotrofos/citología , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Hormona Luteinizante/genética , Masculino , Oryzias/genética , Maduración Sexual/efectos de los fármacos , Maduración Sexual/genética
11.
Biol Reprod ; 102(1): 156-169, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31504222

RESUMEN

Gonadotropes represent approximately 5-15% of the total endocrine cell population in the mammalian anterior pituitary. Therefore, assessing the effects of experimental manipulation on virtually any parameter of gonadotrope biology is difficult to detect and parse from background noise. In non-rodent species, applying techniques such as high-throughput ribonucleic acid (RNA) sequencing is problematic due to difficulty in isolating and analyzing individual endocrine cell populations. Herein, we exploited cell-specific properties inherent to the proximal promoter of the human glycoprotein hormone alpha subunit gene (CGA) to genetically target the expression of a fluorescent reporter (green fluorescent protein [GFP]) selectively to ovine gonadotropes. Dissociated ovine pituitary cells were cultured and infected with an adenoviral reporter vector (Ad-hαCGA-eGFP). We established efficient gene targeting by successfully enriching dispersed GFP-positive cells with flow cytometry. Confirming enrichment of gonadotropes specifically, we detected elevated levels of luteinizing hormone (LH) but not thyrotropin-stimulating hormone (TSH) in GFP-positive cell populations compared to GFP-negative populations. Subsequently, we used next-generation sequencing to obtain the transcriptional profile of GFP-positive ovine gonadotropes in the presence or absence of estradiol 17-beta (E2), a key modulator of gonadotrope function. Compared to non-sorted cells, enriched GFP-positive cells revealed a distinct transcriptional profile consistent with established patterns of gonadotrope gene expression. Importantly, we also detected nearly 200 E2-responsive genes in enriched gonadotropes, which were not apparent in parallel experiments on non-enriched cell populations. From these data, we conclude that CGA-targeted adenoviral gene transfer is an effective means for selectively labeling and enriching ovine gonadotropes suitable for investigation by numerous experimental approaches.


Asunto(s)
Estradiol/farmacología , Gonadotrofos/efectos de los fármacos , Adenohipófisis/efectos de los fármacos , Adenoviridae , Animales , Gonadotrofos/metabolismo , Hormona Luteinizante/metabolismo , Adenohipófisis/metabolismo , Ovinos , Tirotropina/metabolismo
12.
Gen Comp Endocrinol ; 287: 113340, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31778712

RESUMEN

In goldfish, two native isoforms of gonadotropin-releasing hormone (GnRH2 and GnRH3) stimulate luteinizing hormone (LH) and growth hormone (GH) release from pituitary cells through activation of cell-surface GnRH-receptors (GnRHRs) on gonadotrophs and somatotrophs. Interestingly, GnRH2 and GnRH3 induce LH and GH release via non-identical post-receptor signal transduction pathways in a ligand- and cell-type-selective manner. In this study, we examined the involvement of ß-arrestins in the control of GnRH-induced LH and GH secretion from dispersed goldfish pituitary cells. Treatment with Barbadin, which interferes with ß-arrestin and ß2-adaptin subunit interaction, reduced LH responses to GnRH2 and GnRH3, as well as GH responses to GnRH2; but enhanced GnRH3-induced GH secretion. Barbadin also had positive influences on basal hormone release, and basal GH release in particular, as well as basal activity of extracellular signal-regulated kinase (ERK) and GnRH-induced ERK activation. These findings indicate that ß-arrestins play permissive roles in the control of GnRH-stimulated LH release. However, in somatotrophs, ß-arrestins, perhaps by mediating agonist-selective endosomal trafficking of engaged GnRHRs, participate in GnRH-isoform-specific GH release responses (stimulatory and inhibitory for GnRH2-GnRHR and GnRH3-GnRHR activation, respectively). The correlative stimulatory influences of Barbadin on basal hormone release and ERK activation suggest that ß-arrestins may negatively regulate basal secretion through modulation of basal ERK activity. These results provide the first direct evidence of a role for ß-arrestins in hormone secretion from an untransformed primary pituitary cell model, and establish these proteins as important receptor-proximal players in mediating functional selectivity downstream of goldfish GnRHRs.


Asunto(s)
Carpa Dorada , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Somatotrofos/efectos de los fármacos , beta-Arrestinas/fisiología , Animales , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Carpa Dorada/metabolismo , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hormona del Crecimiento/metabolismo , Hormona Luteinizante/metabolismo , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Somatotrofos/metabolismo , beta-Arrestinas/antagonistas & inhibidores
13.
Cells ; 8(9)2019 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-31540096

RESUMEN

C-type natriuretic peptide (CNP) is the most conserved member of the mammalian natriuretic peptide family, and is implicated in the endocrine regulation of growth, metabolism and reproduction. CNP is expressed throughout the body, but is particularly abundant in the central nervous system and anterior pituitary gland. Pituitary gonadotropes are regulated by pulsatile release of gonadotropin releasing hormone (GnRH) from the hypothalamus, to control reproductive function. GnRH and CNP reciprocally regulate their respective signalling pathways in αT3-1 gonadotrope cells, but effects of pulsatile GnRH stimulation on CNP expression has not been explored. Here, we examine the sensitivity of the natriuretic peptide system in LßT2 and αT3-1 gonadotrope cell lines to continuous and pulsatile GnRH stimulation, and investigate putative CNP target genes in gonadotropes. Multiplex RT-qPCR assays confirmed that primary mouse pituitary tissue express Nppc,Npr2 (encoding CNP and guanylyl cyclase B (GC-B), respectively) and Furin (a CNP processing enzyme), but failed to express transcripts for Nppa or Nppb (encoding ANP and BNP, respectively). Pulsatile, but not continuous, GnRH stimulation of LßT2 cells caused significant increases in Nppc and Npr2 expression within 4 h, but failed to alter natriuretic peptide gene expression in αT3-1 cells. CNP enhanced expression of cJun, Egr1, Nr5a1 and Nr0b1, within 8 h in LßT2 cells, but inhibited Nr5a1 expression in αT3-1 cells. Collectively, these data show the gonadotrope natriuretic peptide system is sensitive to pulsatile GnRH signalling, and gonadotrope transcription factors are putative CNP-target genes. Such findings represent additional mechanisms by which CNP may regulate reproductive function.


Asunto(s)
Gonadotrofos/metabolismo , Péptido Natriurético Tipo-C/metabolismo , Células Cultivadas , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Humanos , Péptido Natriurético Tipo-C/genética
14.
Biol Reprod ; 101(4): 791-799, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31290547

RESUMEN

The immortalized mouse gonadotrope cell lines alphaT3-1 and LbetaT2 cells have been a substitute model for primary gonadotropes. These cell lines have provided a homogeneous cell population, as compared to the dissociated anterior pituitaries, which contain a heterogeneous population of cells potentially responsive to estradiol-17beta (E2). Nonclassical actions of E2 assumed to occur through the plasma membrane estrogen receptor 1 (ESR1, also known as ERalpha). These actions have included inhibition of gonadotropin-releasing hormone (GnRH)-induced increases in intracellular calcium concentrations and phosphorylation of p44/42 mitogen-activated protein kinase (ERK-1/2) in ovine pituitaries including primary gonadotropes in vitro. The objective of the present experiment was to determine if alphaT3-1 and LbetaT2 are cell models with limitations to examine the nonclassical actions of E2 occurring in gonadotropes. Experiments were conducted to determine if the cells have ESR1 at the plasma membrane using biotinylation cell and isolation of surface protein and staining with a fluorescently labeled E2 conjugate. The alphaT3-1 cells contain ESR1 associated with but not enriched within lipid rafts of the plasma membrane and do not translocate to lipid rafts upon binding of E2. In contrast, LbetaT2 cells lack ESR1 associated with the plasma membrane. Pretreatment with E2 did not cause inhibition of GnRH-stimulated increases in intracellular concentrations of calcium for either cell type. Phosphorylation of ERK-1/2 was not stimulated by E2 in either cell type. Although these cells lines have been used extensively to study GnRH signaling, in vitro or in vivo effects of nonclassical actions of E2 cannot be replicated in either cell line.


Asunto(s)
Estradiol/farmacología , Gonadotrofos/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Línea Celular Transformada , Gonadotrofos/citología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Adenohipófisis/citología , Adenohipófisis/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de los fármacos
15.
J Reprod Dev ; 65(4): 381-388, 2019 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-31006726

RESUMEN

Cyclic adenosine monophosphate (cAMP) plays a pivotal role in gonadotrope responses in the pituitary. Gonadotropin-releasing hormone (GnRH) mediated synthesis and secretion of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) are regulated by both the Gs/cAMP and Gq/Ca2+ signaling pathways. Pituitary adenylate cyclase-activating polypeptide (PACAP) also regulates GnRH responsiveness in gonadotropes through the PACAP receptor, which activates the Gs/cAMP signaling pathway. Therefore, measuring intracellular cAMP levels is important for elucidating the molecular mechanisms of FSH and LH synthesis and secretion in gonadotropes. The GloSensor cAMP assay is useful for detecting cAMP levels in intact, living cells. In this study, we found that increased GloSensor luminescence intensity did not correlate with cAMP accumulation in LßT2 cells under low pH conditions. This result indicates that cell type and condition must be considered when using GloSensor cAMP.


Asunto(s)
Bioensayo/métodos , AMP Cíclico/análisis , AMP Cíclico/metabolismo , Gonadotrofos/metabolismo , Mediciones Luminiscentes , Animales , Técnicas Biosensibles/métodos , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Hormona Folículo Estimulante/metabolismo , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Luminiscencia , Hormona Luteinizante/metabolismo , Ratones , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
16.
Mol Cell Endocrinol ; 492: 110438, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31034837

RESUMEN

Gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH), are heterodimers of a common α subunit and unique ß subunits. Regulation of their levels, primarily by GnRH, is critical for reproductive function. Several other hormones modulate gonadotropin expression, either independently or by modifying the responsiveness to GnRH. Pituitary adenylate cyclase activating peptide (PACAP) is one such hormone. Four-hour treatment of female mouse primary pituitary cells by either GnRH or PACAP induced FSHß expression, while 24-h treatment repressed FSHß. Both PACAP and GnRH caused FSH secretion into the medium. In the gonadotropes, PACAP activates primarily Gαs and increases concentration of cAMP, while GnRH primarily functions via Gαq and increases calcium concentration. Herein, we compared PACAP and GnRH signaling pathways that lead to the induction of FSHß expression. Interestingly, constitutively active Gαs represses LHß and induces FSHß expression, while Gαq induces both ß-subunits. We determined that FSHß induction by PACAP requires functional EPAC, a cAMP sensor protein that serves as a guanine exchange factors for small G proteins that then bridges cAMP signaling to MAPK pathway. We further demonstrate that in addition to the prototypical small G protein Ras, two members of the Rho subfamily, Rac and CDC42 are also necessary for PACAP induction of FSHß, likely via activation of p38 MAPK that leads to induction of cFOS, a critical transcription factor that is necessary and sufficient for FSHß induction. Therefore, PACAP-induced cAMP pathway leads to MAPK activation that stimulates cFOS induction, to induce the expression of FSHß subunit and increase FSH concentration.


Asunto(s)
Hormona Folículo Estimulante de Subunidad beta/genética , Gonadotrofos/citología , Hormona Liberadora de Gonadotropina/farmacología , Factores de Intercambio de Guanina Nucleótido/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Animales , Línea Celular , AMP Cíclico/metabolismo , Femenino , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Gonadotrofos/efectos de los fármacos , Gonadotrofos/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas ras/metabolismo
17.
J Mol Endocrinol ; 62(2): 67-78, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30481159

RESUMEN

Fertility is dependent on follicle-stimulating hormone (FSH), a product of gonadotrope cells of the anterior pituitary gland. Hypothalamic gonadotropin-releasing hormone (GnRH) and intra-pituitary activins are regarded as the primary drivers of FSH synthesis and secretion. Both stimulate expression of the FSH beta subunit gene (Fshb), although the underlying mechanisms of GnRH action are poorly described relative to those of the activins. There is currently no consensus on how GnRH regulates Fshb transcription, as results vary across species and between in vivo and in vitro approaches. One of the more fully developed models suggests that the murine Fshb promoter is tonically repressed by histone deacetylases (HDACs) and that GnRH relieves this repression, at least in immortalized murine gonadotrope-like cells (LßT2 and αT3-1). In contrast, we observed that the class I/II HDAC inhibitor trichostatin A (TSA) robustly inhibited basal, activin A-, and GnRH-induced Fshb mRNA expression in LßT2 cells and in primary murine pituitary cultures. Similar results were obtained with the class I specific HDAC inhibitor, entinostat, whereas two class II-specific inhibitors, MC1568 and TMP269, had no effects on Fshb expression. Collectively, these data suggest that class I HDACs are positive, not negative, regulators of Fshb expression in vitro and that, contrary to earlier reports, GnRH may not stimulate Fshb by inhibiting HDAC-mediated repression of the gene.


Asunto(s)
Hormona Folículo Estimulante de Subunidad beta/metabolismo , Gonadotrofos/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , 17-Hidroxiesteroide Deshidrogenasas/metabolismo , Activinas/metabolismo , Animales , Línea Celular , Células Cultivadas , Proteína Forkhead Box L2/metabolismo , Gonadotrofos/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Ratones , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo
18.
Histochem Cell Biol ; 151(4): 291-303, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30511269

RESUMEN

Diethylstilbestrol (DES), an estrogen agonist, increases prolactin (PRL) cells through transdifferentiation of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) cells to PRL cells as well as proliferation of PRL cells in adult male mouse pituitary. Since hyperacetylation of histone H3 is implicated in the regulation of activation of various genes, we examined the effect of DES on the state of histone H3 acetylation. DES significantly reduced the immunohistochemical signal for acetylated histone H3 at lysine 9 (H3K9ac) in PRL, LH and FSH cells, but not for H3K18ac or H3K23ac. DES-treated mice were injected intraperitoneally with HDAC inhibitors (HDACi), sodium phenylbutyrate (NaPB) or valproic acid (VPA), to mimic the acetylation level of histone H3. As expected, HDACi treatment restored the level of H3K9ac expression in these cells, and also inhibited DES-induced increase in PRL cells. Furthermore, NaPB and VPA also abrogated the effects of DES on the population density of both LH and FSH cells. Similarly, the numbers of proliferating and apoptotic cells in the pituitary in NaPB- or VPA-treated mice were comparable to those of the control mice. Considered together, these results indicated that the acetylation level of histone H3 plays an important role in DES-induced transdifferentiation of LH to PRL cells as well as proliferation of PRL cells.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Gonadotrofos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Lactotrofos/efectos de los fármacos , Fenilbutiratos/farmacología , Hipófisis/efectos de los fármacos , Ácido Valproico/farmacología , Acetilación/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dietilestilbestrol/administración & dosificación , Dietilestilbestrol/farmacología , Gonadotrofos/citología , Inhibidores de Histona Desacetilasas/administración & dosificación , Histonas/análisis , Histonas/biosíntesis , Inyecciones Intraperitoneales , Lactotrofos/citología , Masculino , Ratones , Ratones Endogámicos ICR , Fenilbutiratos/administración & dosificación , Hipófisis/metabolismo , Conejos , Ácido Valproico/administración & dosificación
19.
Neuroendocrinology ; 108(2): 65-83, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30368511

RESUMEN

BACKGROUND/OBJECTIVES: Anti-Müllerian hormone (AMH) signaling is critical for sexual differentiation and gonadal function. AMH receptor type 2 (AMHR2) is expressed in extragonadal sites such as brain, and pituitary and emerging evidence indicates that AMH biological action is much broader than initially thought. We recently reported that AMH signaling enhances follicle-stimulating hormone synthesis in pituitary gonadotrope cells. However, mechanisms regulating AMHR2 expression in these extragonadal sites remain to be explored. METHOD/RESULTS: Here, we demonstrated in perifused murine LßT2 gonadotrope cells that Amhr2 expression is differentially regulated by GnRH pulse frequency with an induction under high GnRH pulsatility. Furthermore, we showed that GnRH transactivates the human AMHR2 promoter in LßT2 cells. Successive deletions of the promoter revealed the importance of a short proximal region (-53/-37 bp) containing an Egr1 binding site. Using site-directed mutagenesis of Egr1 motif and siRNA mediated-knockdown of Egr1, we demonstrated that Egr1 mediates basal and GnRH-dependent activity of the promoter, identifying Egr1 as a new transcription factor controlling hAMHR2 expression. We also showed that SF1 and ß-catenin are required for basal promoter activity and demonstrated that both factors contribute to the GnRH stimulatory effect, independently of their respective binding sites. Furthermore, using a constitutively active mutant of FOXO1, we identified FOXO1 as a negative regulator of basal and GnRH-dependent AMHR2 expression in gonadotrope cells. CONCLUSIONS: This study identifies GnRH as a regulator of human AMHR2 expression, further highlighting the importance of AMH signaling in the regulation of gonadotrope function.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína Forkhead Box O1/metabolismo , Gonadotrofos/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Receptores de Péptidos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Animales , Línea Celular , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína Forkhead Box O1/genética , Regulación de la Expresión Génica , Gonadotrofos/metabolismo , Ratones , Regiones Promotoras Genéticas , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética
20.
J Endocrinol ; 240(2): 361-377, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30594119

RESUMEN

Luteinizing hormone (Lh) and follicle-stimulating hormone (Fsh) control reproduction in vertebrates. Using a transgenic line of medaka, in which green fluorescent protein expression is controlled by the endogenous lhb promotor, we studied development and plasticity of Lh cells, comparing juveniles and adults of both genders. Confocal imaging and 3D reconstruction revealed hypertrophy and hyperplasia of Lh cells in both genders from juvenile to adult stages. We show that Lh cell hyperplasia may be caused by recruitment of existing pituitary cells that start to produce lhb, as evidenced by time lapse recordings of primary pituitary cell cultures, and/or through Lh cell proliferation, demonstrated through a combination of 5-bromo-2'-deoxyuridine incubation experiments and proliferating cell nuclear antigen staining. Proliferating Lh cells do not belong to the classical type of multipotent stem cells, as they do not stain with anti-sox2. Estradiol exposure in vivo increased pituitary cell proliferation, particularly Lh cells, whereas pituitary lhb and gpa expression levels decreased. RNA-seq and in situ hybridization showed that Lh cells express two estrogen receptors, esr1 and esr2b, and the aromatase gene cyp19a1b, suggesting a direct effect of estradiol, and possibly androgens, on Lh cell proliferation. In conclusion, our study reveals a high degree of plasticity in the medaka Lh cell population, resulting from a combination of recruitment and cell proliferation.


Asunto(s)
Plasticidad de la Célula/fisiología , Proliferación Celular/fisiología , Gonadotrofos/metabolismo , Hipófisis/citología , Factores de Edad , Animales , Animales Modificados Genéticamente , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Hormona Folículo Estimulante/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Gonadotrofos/efectos de los fármacos , Hormona Luteinizante/metabolismo , Masculino , Microscopía Confocal , Oryzias/genética , Oryzias/crecimiento & desarrollo , Oryzias/metabolismo , Imagen de Lapso de Tiempo/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA