Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Biol Reprod ; 96(5): 1043-1051, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28863434

RESUMEN

We examined direct effect of kisspeptin on pituitary gonadotrophs. Kisspeptin-10 (KP10) significantly increased the promoter activities of the gonadotropin subunits, common alpha-glycoprotein (Cga), luteinizing hormone beta (Lhb), and follicle-stimulatinghormone beta (Fshb) in LbetaT2 cells overexpressing kisspeptin receptor (Kiss1r). KP10 and gonadotropin-releasing hormone (GnRH) increased gonadotropin subunit levels to similar degrees and combined treatment with GnRH and KP10 did not potentiate their individual effects. Adenylate cyclase-activating polypeptide 1 (ADCYAP1) also stimulates all three gonadotropin subunits. When cells were stimulated with both KP10 and ADCYAP1, expression of gonadotropin subunits was further increased compared to KP10 or ADCYAP1 alone. KP10 and GnRH dramatically increased serum response element (Sre) promoter levels but only slightly increased cAMP response element (Cre) promoter levels. Combined stimulation with KP10 and GnRH further increased Sre promoter levels. In contrast, ADCYAP1 slightly increased Sre promoter expression but did not modify the effect of KP10. However, ADCYAP1 increased Cre promoter to greater levels than KP10 alone, and combined treatment with KP10 and ADCYAP1 further increased Cre promoter expression. KP10 increased the expression of ADCYAP1 type I receptor (Adcyap1r) and the basal activity of the Cga promoter was increased at a higher Adcyap1r transfection level. The KP10-induced fold increase in all three gonadotropin subunit promoters was not altered by transfection with a higher amount of Adcyap1r vector. Our findings using model cells show that distinct signaling activation by ADCYAP1 potentiates the action of KP10. We also found that KP10 increases Adcyap1r expression.


Asunto(s)
Kisspeptinas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Animales , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Regulación de la Expresión Génica/genética , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Kisspeptinas/genética , Hormona Luteinizante de Subunidad beta/metabolismo , Ratones , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Plásmidos , Regiones Promotoras Genéticas , Receptores de Kisspeptina-1/genética , Elemento de Respuesta al Suero/genética
2.
Biol Reprod ; 97(2): 240-248, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-29044434

RESUMEN

Feeding-related metabolic factors exert regulatory influences on the hypothalamic-pituitary-gonadal axis. Glucagon-like peptide-1 (GLP-1) is an anorexigenic hormone synthesized from the ileum in response to food intake. The purpose of this study was to examine the direct effect of GLP-1 on hypothalamic kisspeptin and gonadotropin-releasing hormone (GnRH) expression using the rat clonal hypothalamic cell line rHypoE-8. GLP-1 significantly increased Kiss-1 mRNA expression in rHypoE-8 cells up to 1.94 ± 0.22-fold. This effect of GLP-1 on Kiss-1 gene expression was also observed in GT1-7 GnRH-producing neurons and in primary cultures of fetal rat brain. GLP-1 increased cAMP-mediated signaling, as determined by cAMP response element activity assays, but failed to activate extracellular signal-regulated kinase pathways. Another anorexigenic factor, leptin, similarly increased Kiss-1 mRNA levels up to 1.34 ± 0.08-fold in rHypoE-8 cells. However, combined treatment with GLP-1 and leptin failed to potentiate their individual effects on Kiss-1 mRNA expression. Gnrh mRNA expression was not significantly increased by GLP-1 stimulation in rHypoE-8, but kisspeptin significantly stimulated the expression of Gnrh mRNA in these cells. Our current observations suggest that the anorexigenic peptide GLP-1 directly regulates Kiss-1 mRNA expression in these hypothalamic cell lines and in neuronal cells of fetal rat brain and affects the expression of Gnrh mRNA.


Asunto(s)
Péptido 1 Similar al Glucagón/farmacología , Kisspeptinas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Animales , Línea Celular , Femenino , Feto/citología , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/citología , Kisspeptinas/genética , Leptina/farmacología , Ratas
3.
Gen Comp Endocrinol ; 247: 138-142, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28131616

RESUMEN

Hypothalamic secretion of gonadotropin-releasing hormone (GnRH) has been established as a principle pathway for initiating and integrating female reproductive function. GnRH stimulates the release of two gonadotropins-luteinizing hormone and follicle-stimulating hormone-from the anterior pituitary, which eventually stimulate the synthesis of sex steroids in association with follicular growth and ovulation. This reproductive control of the hypothalamic-pituitary-gonadal (HPG) axis also mediates gonadal feedback mechanisms. Although GnRH neurons certainly play a pivotal role in the HPG axis, the detailed mechanisms of their functional network, including regulatory systems, remain unknown. After the discovery of the indispensable role of kisspeptin in the development of human reproductive functions, our understanding of the neuroendocrine regulation of the HPG axis was revolutionized, and it is now recognized that kisspeptin acts upstream of GnRH and is responsible for sex steroid feedback mechanisms. Kisspeptin can stimulate gonadotropin release from the pituitary gland by stimulating GnRH release and GnRH antagonists prevent kisspeptin-induced gonadotropin release. Furthermore, it has been shown that GnRH neurons express kisspeptin receptors. Nevertheless, the detailed mechanisms underlying the regulation of homogeneous populations of GnRH neurons are still largely unknown because of the limitations of experimental models used for investigation. The hypothalamus consists of a complex network of distinct neuronal cells, and it is difficult to isolate single-cell populations of GnRH neurons. The establishment of GnRH-expressing cell lines has allowed us to examine the events happening at the single-cell level. In this review, we describe in vitro studies using a GnRH-producing cell model, GT1-7 cells, which have been used to examine how GnRH-producing cells respond to hypothalamic factors and how they are involved in GnRH synthesis.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Modelos Biológicos , Neuronas/metabolismo , Animales , Línea Celular , Kisspeptinas/metabolismo
4.
Gen Comp Endocrinol ; 246: 382-389, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28087300

RESUMEN

Hypothalamic kisspeptin plays a pivotal role in the regulation of the hypothalamic-pituitary-gonadal axis by stimulating gonadotropin-releasing hormone (GnRH) release into the portal circulation, with the subsequent release of gonadotropins. Kisspeptin and its receptor, the kisspeptin 1 receptor (Kiss1R), are also expressed in the pituitary gland. This study demonstrates the interaction between GnRH and kisspeptin within the pituitary gonadotrophs by altering their individual receptor expression. Our results show that kisspeptin and Kiss1R are expressed in the mouse pituitary gonadotroph cell line LßT2. Endogenous Kiss1R did not respond to kisspeptin and failed to stimulate gonadotropin LHß and FSHß expression in LßT2 cells; however, kisspeptin increased both LHß and FSHß promoter activity in Kiss1R-overexpressing LßT2 cells. Stimulating the cells with GnRH significantly increased Kiss1R expression, whereas kisspeptin increased the expression of the GnRH receptor (GnRHR) in these cells. Elevating the Kiss1R concentration led to an increase in the basal activities of gonadotropin LHß- and FSHß-subunit promoters. In addition, the level of kisspeptin-induced LHß promoter activity, but not that of FSHß, was significantly increased when a large number of Kiss1R expression vectors was introduced into the cells. The level of induction of GnRH-induced gonadotropin promoter activities was not significantly changed by increasing Kiss1R expression. Increasing the amount of GnRHR by overexpressing cellular GnRHR did not potentiate basal gonadotropin promoter activities; however, kisspeptin- and GnRH-stimulated increases in gonadotropin promoter activities were significantly potentiated (except GnRH-induced LHß promoters). The activities of serum response element-containing promoters were also modified in cells overexpressing Kiss1R or GnRHR. Our current observations demonstrate that GnRH and kisspeptin affect each other's function to stimulate gonadotropin subunit gene expression by reciprocally increasing the expression of their receptors.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Gonadotrofos/metabolismo , Kisspeptinas/metabolismo , Receptores LHRH/metabolismo , Animales , Ratones
5.
Gen Comp Endocrinol ; 242: 59-65, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26654743

RESUMEN

The selective histone deacetylase inhibitor, trichostatin A (TSA), increases follicle-stimulating hormone ß subunit (FSHß) mRNA expression but not α- and luteinizing hormone ß (LHß)-subunits in both the pituitary gonadotrophic cell line LßT2 and primary cultures of rat anterior pituitary cells. TSA increased histone acetylation in whole cell lysates in both cells. In addition, retinaldehyde dehydrogenases (RALDHs), which are retinoic acid (RA)-synthesizing enzymes, were induced by TSA in these cells. Anacardic acid, a histone acetyltransferase inhibitor that prevents histone acetylation, significantly inhibited TSA-induced FSHß mRNA expression as well as TSA-induced RALDH2 and RALDH3 mRNA expression. Similar to the effect of TSA, gonadotropin-releasing hormone stimulated RALDH expression in LßT2 cells. RA directly applied to the pituitary cells stimulated the transcriptional activity of the FSHß promoter. In addition, α- and LHß-subunit promoters were also activated by RA. Our results suggest that TSA specifically increases FSHß expression with a concomitant increase in RALDHs; however, RALDH and RA are not directly involved in the specific regulation of FSHß by TSA.


Asunto(s)
Hormona Folículo Estimulante de Subunidad beta/metabolismo , Regulación de la Expresión Génica/fisiología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Retinal-Deshidrogenasa/metabolismo , Tretinoina/metabolismo , Acetilación , Animales , Hormona Folículo Estimulante/metabolismo , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Inhibidores de Histona Desacetilasas , Hormona Luteinizante de Subunidad beta/metabolismo , Hipófisis/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Ratas , Retinal-Deshidrogenasa/genética , Activación Transcripcional/efectos de los fármacos
6.
Int J Mol Sci ; 17(10)2016 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-27681724

RESUMEN

Gonadotropin-releasing hormone (GnRH) and gonadotropins are indispensable hormones for maintaining female reproductive functions. In a similar manner to other endocrine hormones, GnRH and gonadotropins are controlled by their principle regulators. Although it has been previously established that GnRH regulates the synthesis and secretion of luteinizing hormone (LH) and follicle-stimulating hormone (FSH)-both gonadotropins-from pituitary gonadotrophs, it has recently become clear that hypothalamic GnRH is under the control of hypothalamic kisspeptin. Prolactin, which is also known as luteotropic hormone and is released from pituitary lactotrophs, stimulates milk production in mammals. Prolactin is also regulated by hypothalamic factors, and it is thought that prolactin synthesis and release are principally under inhibitory control by dopamine through the dopamine D2 receptor. In addition, although it remains unknown whether it is a physiological regulator, thyrotropin-releasing hormone (TRH) is a strong secretagogue for prolactin. Thus, GnRH, LH and FSH, and prolactin are mainly regulated by hypothalamic kisspeptin, GnRH, and TRH, respectively. However, the synthesis and release of these hormones is also modulated by other neuropeptides in the hypothalamus. Pituitary adenylate cyclase-activating polypeptide (PACAP) is a hypothalamic peptide that was first isolated from sheep hypothalamic extracts based on its ability to stimulate cAMP production in anterior pituitary cells. PACAP acts on GnRH neurons and pituitary gonadotrophs and lactotrophs, resulting in the modulation of their hormone producing/secreting functions. Furthermore, the presence of the PACAP type 1 receptor (PAC1R) has been demonstrated in these cells. We have examined how PACAP and PAC1R affect GnRH- and pituitary hormone-secreting cells and interact with their principle regulators. In this review, we describe our understanding of the role of PACAP and PAC1R in the regulation of GnRH neurons, gonadotrophs, and lactotrophs, which are regulated mainly by kisspeptin, GnRH, and TRH, respectively.

7.
Biol Reprod ; 92(2): 45, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25519184

RESUMEN

4-Chloro-N-[6,8-dibromo-2-(2-thienyl)imidazo[1,2-alpyridine-3-yl] (DS1) is a GABA(A) receptor agonist that selectively binds to delta subunit-containing GABA(A) alpha4beta3delta receptors. In the present study, we examined the effect of DS1 on pituitary gonadotropin subunit gene expression using the mouse pituitary gonadotroph cell line LbetaT2. DS1 increased the promoter activity of the gonadotropin subunits luteinizing hormone beta (LHbeta), follicle-stimulating hormone beta (FSHbeta), and alpha. Gonadotropin-releasing hormone (GnRH) receptor promoters were also activated by DS1. The effects of DS1 on gonadotropin subunit promoters were obvious, but they were less than those induced by stimulation with GnRH. GnRH-stimulated gonadotropin subunit promoters were enhanced in the presence of DS1. A prototypic specific agonist for GABAA receptors, muscimol, failed to increase LHbeta and FSHbeta subunit promoter activity and had no effect on GnRH-increased LHbeta and FSHbeta promoter activity. In addition, SKF97541, a specific agonist for GABAB receptors, did not modulate basal or GnRH-induced LHbeta and FSHbeta promoter activity. A natural GABA compound failed to increase gonadotropin promoter activity and potentiated the effect of GnRH on the FSHbeta promoter. DS1 increased the activity of serum response element (SRE) and cAMP response element (CRE) promoters, which reflect the activity of the extracellular signal-regulated kinase and cAMP/protein kinase A (PKA) pathways, and GnRH-increased SRE and CRE promoter activity was enhanced in the presence of DS1. A specific inhibitor of the ERK signaling pathway, U0126, prevented DS1-induced LHbeta and FSHbeta promoter activity almost completely; however, H89, a PKA inhibitor, did not modulate the effect of DS1. Our current observations demonstrate that the GABAA alpha4beta3delta receptor agonist DS1 can stimulate gonadotropin subunit gene expression in association with the ERK signaling pathway.


Asunto(s)
Hormona Folículo Estimulante/genética , Agonistas del GABA/farmacología , Expresión Génica/efectos de los fármacos , Gonadotrofos/efectos de los fármacos , Hormona Luteinizante de Subunidad beta/genética , Animales , Línea Celular , Hormona Folículo Estimulante/metabolismo , Gonadotrofos/metabolismo , Hormona Luteinizante de Subunidad beta/metabolismo , Ratones , Muscimol/farmacología , Regiones Promotoras Genéticas , Subunidades de Proteína/metabolismo , Receptores de GABA-A/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Reprod Biol Endocrinol ; 13: 90, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26268437

RESUMEN

BACKGROUND: Gonadotropin-releasing hormone (GnRH) and kisspeptin in the hypothalamus are thought to be crucial components of the hypothalamic-pituitary-gonadal (HPG) axis and maintain reproductive function. These neuropeptides are also expressed in the placenta, where they may contribute to placental physiology. In this study, we examined how these peptides are regulated within the placenta. METHODS: We used primary cultures of placental tissue from rats of 16-18 days gestation. After stimulation with estradiol, GnRH, kisspeptin, and neurokinin B (NKB), changes in placental GnRH, kisspeptin, and human chorionic gonadotropin (hCG) mRNA expression were evaluated by real-time quantitative RT-PCR analysis. RESULTS: Immunocytochemical analysis showed that rat placental cells contained cells expressing kisspeptin or GnRH. GnRH and kisspeptin mRNA expression was significantly increased in placental cells in the presence of estradiol; NKB mRNA expression was also stimulated by estradiol. Stimulation of the cells with kisspeptin failed to stimulate GnRH mRNA expression. Conversely, both GnRH itself and NKB increased GnRH mRNA expression. Kisspeptin mRNA expression was not increased by kisspeptin itself; however, GnRH and NKB significantly increased kisspeptin mRNA expression. hCG expression was increased in the presence of estradiol. In addition, kisspeptin, GnRH, and NKB could stimulate the expression of hCG mRNA in placental cells. CONCLUSIONS: Our experiments using primary cultures of rat placental cells showed that GnRH, kisspeptin, and NKB expression was enhanced by estradiol, and unlike in the hypothalamus, kisspeptin did not control the expression of GnRH in placental cells. NKB might be located upstream of kisspeptin and GnRH, and these neuropeptides might be involved in the induction of hCG expression in placental cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hormona Liberadora de Gonadotropina/biosíntesis , Kisspeptinas/biosíntesis , Placenta/metabolismo , Animales , Células Cultivadas , Estradiol/farmacología , Femenino , Hormona Liberadora de Gonadotropina/genética , Humanos , Kisspeptinas/genética , Neuroquinina B/biosíntesis , Neuroquinina B/genética , Placenta/citología , Placenta/efectos de los fármacos , Embarazo , Ratas
9.
Endocr J ; 61(4): 335-42, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24430728

RESUMEN

Trichostatin A (TSA) is a selective inhibitor of mammalian histone deacetylase. In the present study, TSA was found to selectively increase gene expression of the pituitary gonadotropin ß-subunit of follicle-stimulating hormone (FSH). Stimulation of mouse pituitary gonadotroph cell lines, LßT2, with TSA for 24 h resulted in no change in mRNA expression of the α- and LHß-subunit. On the other hand, FSHß-subunit mRNA expression was significantly increased in a dose-dependent fashion. Similarly, specific induction of the FSHß-subunit gene with TSA stimulation was observed in primary cultures of rat pituitary cells. Histone acetylation in whole cell lysates of LßT2 cells was significantly increased after TSA treatment, but not gonadotropin-releasing hormone (GnRH) treatment. The effect of TSA on FSHß mRNA expression was prominent compared to that of GnRH; however, TSA-stimulated FSHß mRNA expression was significantly reduced with combined TSA and GnRH treatment. TSA caused a slight increase in extracellular signal-regulated kinase (ERK) phosphorylation, while GnRH-increased ERK phosphorylation was potentiated in the presence of TSA. In addition, TSA, but not GnRH, significantly stimulated gene expression of retinaldehyde dehydrogenase 1 (RALDH1), a retinoic acid (RA) synthesizing enzyme involved in cell differentiation. These findings demonstrate that TSA specifically increases FSHß subunit gene expression with a concomitant increase in whole cell histone acetylation. Moreover, although GnRH is a stimulator of FSHß gene expression, it interfered with the stimulatory effect of TSA on FSHß mRNA expression, without modification of TSA-increased whole cell histone acetylation. This suggests that the mechanisms of TSA and GnRH-induced gonadotropin subunit gene expression are entirely distinct.


Asunto(s)
Antineoplásicos/farmacología , Hormona Folículo Estimulante de Subunidad beta/agonistas , Regulación de la Expresión Génica/efectos de los fármacos , Gonadotrofos/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Acetilación/efectos de los fármacos , Familia de Aldehído Deshidrogenasa 1 , Animales , Antineoplásicos/química , Línea Celular , Células Cultivadas , Femenino , Hormona Folículo Estimulante de Subunidad beta/genética , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Gonadotrofos/citología , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Liberadora de Gonadotropina/metabolismo , Inhibidores de Histona Desacetilasas/química , Histonas/metabolismo , Ácidos Hidroxámicos/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Hipófisis/citología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Retinal-Deshidrogenasa/química , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
Endocr J ; 61(1): 25-34, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24088662

RESUMEN

Gonadotropin-inhibitory hormone (GnIH) was first identified in quail as a novel neurohormone that acts directly on the anterior pituitary to inhibit gonadotropin release. GnIH inhibits not only gonadotropin release from the pituitary gland but also inhibits the release of gonadotropin-releasing hormone (GnRH) from the hypothalamus. In this study, we examined how GnIH receptors were regulated in pituitary gonadotroph cells and GnRH-producing neurons in the hypothalamus. In the mouse pituitary gonadotroph cell line LßT2, GnRH increased expression of the GnIH receptor, G-protein coupled receptor 74 (GPR74). GnRH also stimulated the expression of GPR74 and GPR147 in primary cultures of rat anterior pituitary cells. In addition, when GnRH was administered to LßT2 cells in a pulsatile manner, low frequency GnRH pulse stimulation stimulated GPR74 and GPR147 expression more than did high frequency GnRH pulses. In the mouse hypothalamic GnRH-producing cell line GT1-7, hypothalamic kisspeptin did not significantly increase the expression of GnIH receptors. However, the intermittent administration of kisspeptin to GT1-7 cells significantly increased GPR74 and GPR147 mRNA expression. The overexpression of either constitutively active MEK kinase (MEKK) or protein kinase A (PKA) in LßT2 cells increased the expression of GPR74 mRNA. Conversely, in GT1-7 cells, although the overexpression of either MEKK or PKA failed to stimulate GnIH receptor expression, the combined overexpression of both kinases together increased GPR74 and GPR147 mRNA levels. Our current observations suggest that two central controllers of reproductive function, GnRH and kisspeptin, stimulate the expression of GnIH receptors in pituitary gonadotroph cells and hypothalamic GnRH neurons.


Asunto(s)
Expresión Génica , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/biosíntesis , Hipotálamo/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Animales , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hormona Liberadora de Gonadotropina/administración & dosificación , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Kisspeptinas/farmacología , Quinasas Quinasa Quinasa PAM/genética , Ratones , ARN Mensajero/análisis , Ratas , Receptores de Neuropéptido/genética
11.
Gen Comp Endocrinol ; 194: 94-101, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24055558

RESUMEN

Kisspeptin signaling through its receptor is crucial for many reproductive functions. However, the molecular mechanisms and biomedical significance of the regulation of GnRH neurons by kisspeptin have not been adequately elucidated. In the present study, we found that kisspeptin increases GnRH receptor (GnRHR) expression in a GnRH-producing cell line (GT1-7). Because cellular activity of G protein-coupled receptor 54 (GPR54) and GnRHR was limited in GT1-7 cells, we overexpressed these receptors to clarify receptor function. Using luciferase reporter constructs, the activity of both the serum response element (Sre) promoter, a target for extracellular signal-regulated kinase (ERK), and the cyclic AMP (cAMP) response element (Cre) promoter were increased by kisspeptin. Although GnRH increased Sre promoter activity, the Cre promoter was not significantly activated by GnRH. Kisspeptin, but not GnRH, increased cAMP accumulation in these cells. Kisspeptin also increased the transcriptional activity of GnRHR; however, the effect of GnRH on the GnRHR promoter was limited and not significant. Transfection of GT1-7 cells with constitutively active MEK kinase (MEKK) and protein kinase A (PKA) increased GnRHR expression. In addition, GnRHR expression was further increased by co-overexpression of MEKK and PKA. The Cre promoter, but not the Sre promoter, was also further activated by co-overexpression of MEKK and PKA. GnRH significantly increased the activity of the GnRHR promoter in the presence of cAMP. The present findings suggest that kisspeptin is a potent stimulator of GnRHR expression in GnRH-producing neurons in association with ERK and the cAMP/PKA pathways.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores LHRH/metabolismo , Animales , Línea Celular , Hormona Liberadora de Gonadotropina/genética , Hipotálamo/metabolismo , Kisspeptinas/genética , Ratones , Regiones Promotoras Genéticas/genética , Receptores Acoplados a Proteínas G/genética , Receptores LHRH/genética
12.
Gen Comp Endocrinol ; 193: 95-102, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23899713

RESUMEN

The present study demonstrates the action of pituitary adenylate cyclase-activating polypeptide (PACAP) on gonadotropin-releasing hormone (GnRH)-producing neuronal cells, GT1-7. Because we found the expression levels of PACAP type 1 receptor (PAC1R) to be low in these cells, we transfected them with PAC1R expression vector and observed the outcome. PACAP increased the activity of the serum response element (Sre) promoter, a target of extracellular signal-regulated kinase (ERK), as well as the cAMP response element (Cre) promoter in GT1-7 cells overexpressing PAC1R. We also observed ERK phosphorylation and cAMP accumulation upon PACAP stimulation. PACAP stimulated the promoter activity of GnRH receptor (GnRHR) with increasing levels of GnRHR proteins. Notably, the increase in GnRHR promoter activity from kisspeptin was potentiated in the presence of PACAP. A similar increasing effect of PACAP on the action of kisspeptin was observed for Cre promoter activity. On the other hand, the Sre promoter activated by kisspeptin was inhibited by co-treatment with kisspeptin and PACAP. Likewise, kisspeptin-increased GnRHR promoter activity and Cre promoter activity were both potentiated in the presence of cAMP, whereas the Sre promoter activated by kisspeptin was inhibited in the presence of cAMP. Our observations show that PACAP increases GnRHR expression and stimulates kisspeptin's effect on GnRHR expression in association with the cAMP/PKA signaling pathway in GT1-7 cells overexpressing PAC1R. In addition, PACAP was shown to have an inhibitory effect on ERK-mediated kisspeptin action.


Asunto(s)
Regulación de la Expresión Génica , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores LHRH/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Animales , Línea Celular , AMP Cíclico/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Ratones , Receptores LHRH/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Transducción de Señal/efectos de los fármacos
13.
Gynecol Endocrinol ; 29(6): 583-7, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23656386

RESUMEN

Kisspeptins are known to be the principle regulators of the hypothalamic-pituitary gonadal (HPG) axis. In addition, the role of pituitary adenylate cyclase-activating polypeptide (PACAP) in the regulation of pituitary gonadotropins has been elucidated. We measured plasma concentrations of kisspeptin and PACAP and determined whether the levels of these peptides varied in proportion to circulating gonadotropin levels. Plasma luteinizing hormone (LH) levels were higher in postmenopausal women and in patients with premature ovarian failure (POF) and lower in patients with idiopathic hypogonadotropic hypogonadism (IHH) compared with the LH level in normally menstruating women. Similarly, serum follicle-stimulating hormone levels were higher in postmenopausal women and in patients with POF but lower in pregnant women and patients with IHH compared with normally menstruating women. Plasma levels of kisspeptins were significantly higher in pregnant women compared with normally menstruating women. However, no significant differences were observed in postmenopausal women, patients with POF, and patients with IHH. On the other hand, plasma levels of PACAP were significantly lower in pregnant women, patients with POF, and in IHH patients when compared with normally menstruating women. No significant differences were observed in PACAP concentration between postmenopausal women and in normally menstruating women. Our observations suggest that the serum levels of kisspeptins and PACAP did not correlate with variations in serum gonadotropin levels.


Asunto(s)
Gonadotropinas/sangre , Kisspeptinas/sangre , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/sangre , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Análisis Químico de la Sangre , Femenino , Humanos , Infertilidad Femenina/sangre , Ciclo Menstrual/sangre , Persona de Mediana Edad , Posmenopausia/sangre , Embarazo/sangre , Adulto Joven
14.
Endocr J ; 59(7): 631-40, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22641014

RESUMEN

The role of Neurokinin B (NKB) and Dynorphin A (Dyn) in the regulation of the hypothalamic pituitary axis is an important area of recent investigation. These peptides are critical for the rhythmic release of GnRH, which subsequently stimulates the secretion of the gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). The present study utilized the gonadotroph cell line LßT2 and the somatolactotroph GH3 cell line to examine the possible role of these peptides in pituitary hormone secretion. The NKB receptor (NK3R) and the Dyn receptor (the κ-opiate receptor (KOR)) were both detected in LßT2 cells and GH3 cells. NKB, by itself, failed to increase gonadotropin LHß and FSHß promoter activities and did not modulate the effects of GnRH on gonadotropin promoter activity. In GH3 cells, NKB significantly increased TRH-induced PRL promoter activity although NKB alone did not have an effect on basal PRL promoter activity. Dyn had no effect on gonadotropin promoters alone or in combination with GnRH stimulation. PRL promoters stimulated by TRH were not significantly changed by Dyn. TRH-induced PRL promoter activity was further increased in the presence of higher concentrations of NKB, whereas Dyn did not have a significant effect on the PRL promoter even at a high concentration. In addition, TRH-induced ERK (Extracelluar signal-regulated kinase) activation was enhanced in the presence of NKB. Our current study demonstrated that NKB had a stimulatory effect on PRL expression in a PRL-producing cell, but had no effect on gonadotropin secretion from a gonadotroph cell line.


Asunto(s)
Dinorfinas/fisiología , Gonadotrofos/metabolismo , Lactotrofos/metabolismo , Neuroquinina B/fisiología , Somatotrofos/metabolismo , Animales , Línea Celular , Dinorfinas/genética , Dinorfinas/metabolismo , Dinorfinas/farmacología , Gonadotrofos/efectos de los fármacos , Gonadotropinas/genética , Gonadotropinas/metabolismo , Lactotrofos/efectos de los fármacos , Neuroquinina B/genética , Neuroquinina B/metabolismo , Neuroquinina B/farmacología , Hipófisis/citología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Prolactina/genética , Prolactina/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Ratas , Receptores Opioides/genética , Receptores Opioides/metabolismo , Receptores de Taquicininas/genética , Receptores de Taquicininas/metabolismo , Somatotrofos/efectos de los fármacos , Transfección , Receptor de Nociceptina
15.
Endocr J ; 59(10): 867-79, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22785235

RESUMEN

We examined the effects of sex steroids on prolactin promoter activity in rat somatolactotrophic GH3 cells. Both estradiol (E2) and progesterone (P4) were found to inhibit basal prolactin promoter activity, but to potentiate Thyrotropin-releasing hormone (TRH)-induced prolactin promoter activity. P4 had a greater inhibitory effect on basal prolactin promoter activity than E2, and P4 also potentiated TRH-induced prolactin promoter more potently than E2. Combined treatment with E2 and P4 further increased TRH-induced prolactin promoter activity. E2 and P4 also both reduced basal serum response element (SRE) promoter activity, and increased TRH-induced SRE promoter activity. Combination treatment with E2 and P4 reduced basal activity of SRE promoter and increased TRH-induced SRE activity more potently than E2 or P4 alone. In contrast, basal cAMP response element (CRE) promoter activity was not influenced by either E2 or P4, although TRH-induced CRE promoter was potentiated by each of these steroids, and was further increased by E2 and P4 combination treatment. Both E2 and P4 increased TRH-induced extracellular signal-regulated kinase (ERK) phosphorylation; however, intracellular cAMP levels was not influenced by E2 or P4. TRH-induced CRE promoter was inhibited by mitogen-activated protein kinase/ERK kinase (MEK) inhibitor and was increased by overexpression of MEK kinase (MEKK). This study showed that ERK and SRE transcriptional pathways, but not the cAMP/CRE pathway, may be involved in the suppression of basal prolactin promoter activity, whereas both the ERK/SRE and MAP kinase-mediated CRE pathways appear to be involved in the increased transcriptional efficiency of the prolactin promoter induced by TRH stimulation.


Asunto(s)
Estradiol/farmacología , Lactotrofos/metabolismo , Progesterona/farmacología , Prolactina/biosíntesis , Animales , Línea Celular Tumoral , AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Ratas , Elemento de Respuesta al Suero/genética , Elemento de Respuesta al Suero/fisiología , Hormona Liberadora de Tirotropina/farmacología
16.
Neuro Endocrinol Lett ; 33(6): 608-13, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23160221

RESUMEN

OBJECTIVES: Sex steroid hormones play roles in the regulation of pituitary hormone synthesis and secretion. Here we investigated the role of estradiol (E2) and progesterone (P4) on pituitary gonadotropin luteinizing hormone (LH)ß- and follicle stimulating hormone (FSH)ß-transcriptional activity in a single colony of gonadotroph LßT2 cells. METHODS: Pituitary gonadotroph cell line, LßT2 cells were used in this study. Cells were transfected with LHß- or FSHß-subunit promoter region-linked luciferase vector, and stimulated with gonadotropin-releasing hormone (GnRH) in the presence or absence of sex steroids. Transcriptional activity for LHß- and FSHß-subunit were determined by luciferase assay. Effects of sex steroids on cell proliferation was also determined by measurement of 5-bromoe-2'-deoxyuridine (BrdU) incorporation. RESULTS: The basal promoter activity of the LHß subunit was not modulated by 10 nM E2, but gonadotropin releasing hormone (GnRH)-induced LHß promoter activity was significantly increased by the same concentration of E2. Similarly, although the basal FSHß promoter was not modulated by 10 nM E2, GnRH-induced FSHß promoters were significantly potentiated in the presence of E2. One micromole E2 modulated neither basal nor GnRH-induced LHß and FSHß promoters. On the other hand, basal LHß promoter activity was enhanced by 1 µM P4, but the stimulatory response of GnRH on LHß promoters was significantly inhibited in the presence of 1 µM P4. Similar to LHß promoters, the basal activity of the FSHß promoter was increased by 1 µM P4; however, the response to GnRH was not modulated in the presence of P4. Ten micromoles P4 modified neither basal nor GnRH-induced promoter activity for LHß and FSHß. E2 had no antagonistic effect on P4-induced basal promoter activities of LHß or FSHß. A cell proliferation assay showed that neither E2 nor P4 modulated the growth of LßT2 cells, even in the presence or absence of GnRH. CONCLUSION: These observations suggest that both E2 and P4 uniquely modulate basal and GnRH-stimulated gonadotropin promoters without affecting cell growth.


Asunto(s)
Estradiol/metabolismo , Hormona Folículo Estimulante de Subunidad beta/genética , Hormona Luteinizante de Subunidad beta/genética , Hipófisis/fisiología , Progesterona/metabolismo , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , Estradiol/farmacología , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Humanos , Luciferasas/genética , Hormona Luteinizante de Subunidad beta/metabolismo , Hipófisis/citología , Hipófisis/efectos de los fármacos , Progesterona/farmacología , Regiones Promotoras Genéticas/fisiología , Transfección
17.
Biochem Biophys Res Commun ; 410(2): 295-300, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21658369

RESUMEN

We examined the effect of the pituitary adenylate cyclase-activating polypeptide (PACAP) type 1 receptor (PAC1R) on gonadotropin-releasing hormone (GnRH)-induced gonadotropin subunit promoter activities using the LßT2 gonadotroph cell line. In mock transfected cells, GnRH-increased LHß and FSHß promoters up to 2.74 ± 0.15-fold and 1.6 ± 0.05-fold respectively. When cells were transfected with PAC1R, both LHß and FSHß promoter activities were further increased up to 6.1 ± 0.87-fold and 2.22 ± 0.43-fold following GnRH stimulation. ERK phosphorylation, serum response element (SRE) promoters, and cAMP response element (CRE) promoters stimulated by GnRH were also potentiated in the presence of increasing amounts of PAC1R. The EC50 values for LHß and FSHß gene transcription by GnRH were significantly decreased by overexpression of PAC1R. PACAP 6-38, a PACAP receptor antagonist, failed to reduce the effect of GnRH on gonadotropin promoter activities in PAC1R overexpressing cells, suggesting that the potentiation of the effects of GnRH by PAC1R expression was not related to an autocrine mechanism of PACAP produced in the gonadotrophs. Our current results show that the action of GnRH in the regulation of gonadotropin subunit expression is enhanced by the presence of PAC1Rs.


Asunto(s)
Regulación de la Expresión Génica , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante de Subunidad beta/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Animales , Línea Celular , Expresión Génica , Ratones , Regiones Promotoras Genéticas
18.
Biol Reprod ; 84(5): 996-1004, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21228211

RESUMEN

In pituitary gonadotrophs, GnRH induces expression of the mitogen-activated protein kinases (MAPK3/1) dephosphorylating enzyme, dual-specificity phosphatase 1 (DUSP1). Here we examined DUSP1 expression levels following pulsatile GnRH stimulation of the LbetaT2 gonadotroph cells. DUSP1 expression was increased more prominently following high-frequency (every 30 min) GnRH pulse stimulation (7.02- ± 1.47-fold) than low-frequency (every 120 min) GnRH pulses (2.68- ± 0.09-fold). With high-frequency GnRH pulses, DUSP1 expression increased by 2.89- ± 0.32-fold 2 h after GnRH pulse initiation (four 5-min pulses). DUSP1 expression was not induced following lower frequency GnRH pulses, even when the GnRH concentration was increased. Under high-frequency conditions, MAPK3/1 phosphorylation was observed 10 min after the GnRH pulse and decreased to basal levels after 25 min. However, MAPK3/1 dephosphorylation did not occur concurrently with DUSP1 expression. Overexpression of MAP3K1, a kinase upstream of MAPK3/1, increased both the Lhb and the Fshb subunit promoter activities, which could be completely inhibited by cotransfection with DUSP1-expressing vectors. Serum response factor (Srf) promoter activities induced by MAP3K1 were also prevented by DUSP1 overexpression, confirming that MAPK3/1 has an important role in gonadotropin subunit gene expression. Both high- and low-frequency GnRH pulse stimulation failed to increase the Lhb and Fshb subunit gonadotropin gene expression levels upon DUSP1 overexpression. Our study demonstrates that DUSP1 is specifically expressed following high-frequency GnRH pulses and that this effect may participate in the differential regulation of gonadotropin subunit expression in association with MAPK3/1 phosphorylation.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/biosíntesis , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante de Subunidad beta/metabolismo , Periodicidad , Hipófisis/metabolismo , Animales , Línea Celular , Inducción Enzimática , Hormona Folículo Estimulante de Subunidad beta/genética , Genes Reporteros , Hormona Luteinizante de Subunidad beta/genética , Quinasa 1 de Quinasa de Quinasa MAP/genética , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Concentración Osmolar , Fosforilación , Regiones Promotoras Genéticas , Ratas , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo
19.
J Dermatol Sci ; 102(3): 158-166, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34049769

RESUMEN

BACKGROUND: The development of BRAF/MEK inhibitors in patients with metastatic melanoma harboring BRAF mutations has garnered attention for liquid biopsy to detect BRAF mutations in cell-free DNA (cfDNA) using droplet digital PCR (ddPCR) or next-generation sequencing methods. OBJECTIVE: To investigate gene mutations in tumor DNA and cfDNA collected from 43 melanoma patients and evaluate their potential as biomarkers. METHODS: ddPCR and CAncer Personalized Profiling by deep Sequencing (CAPP-Seq) techniques were performed to detect gene mutations in plasma cfDNA obtained from patients with metastatic melanoma. RESULTS: Gene variants, including BRAF, NRAS, TP53, GNAS, and MET, were detectable in the plasma cfDNA, and the results were partially consistent with the results of those identified in the tissues. Among the variants examined, copy numbers of MET mutations were consistent with the disease status in two melanoma patients. CONCLUSION: Liquid biopsy using CAPP-Seq and ddPCR has the potential to detect tumor presence and mutations, especially when tissue biopsies are unavailable. MET mutations in cfDNA may be a potential biomarker in patients with metastatic melanoma.


Asunto(s)
Biomarcadores de Tumor/genética , ADN Tumoral Circulante/genética , Melanoma/diagnóstico , Neoplasias Cutáneas/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/sangre , ADN Tumoral Circulante/sangre , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN/métodos , Estudios de Factibilidad , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Biopsia Líquida/métodos , Masculino , Melanoma/sangre , Melanoma/genética , Persona de Mediana Edad , Mutación , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-met/genética , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/genética , Adulto Joven
20.
Eur J Dermatol ; 31(1): 48-54, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33586656

RESUMEN

BACKGROUND: The microbiome plays an important role in the tumour microenvironment (TME). OBJECTIVES: In this study, we investigated the clinical significance of the microbiota in extramammary Paget's disease (EMPD). MATERIALS & METHODS: Patients with EMPD, treated between March 2007 and September 2019 at Kumamoto University Hospital, were investigated retrospectively. Inclusion criteria included: histological diagnosis of EMPD, inspection of the bacterial culture of the cancer lesion using swab sampling, and availability of sufficient tissue in paraffin blocks for immunohistochemistry. For the latter, primary antibodies against IL-17, CD163 and ionized calcium-binding adapter molecule 1 (Iba1) were used. RESULTS: Bacterial cultures of the cancer lesion revealed that Staphylococcus aureus (S. aureus) was highly prevalent in EMPD patients, with dermal invasion or lymph node metastasis, compared to patients without these findings. Furthermore, the number of IL-17-positive cells and CD163-positive M2-like macrophages (pro-tumour macrophages) were increased in EMPD tissues with S. aureus. Moreover, the number of IL-17-producing cells in EMPD tissues positively correlated with the accumulation of CD163-positive M2-like macrophages. In addition, the percentage of CD163-positive cells within Iba-1-positive macrophages (total macrophages) was also significantly elevated in EMPD tissues with S. aureus. CONCLUSION: Based on these findings, S. aureus may exacerbate the pathological condition of EMPD via the accumulation of IL-17 and M2-like macrophages.


Asunto(s)
Interleucina-17/fisiología , Macrófagos/fisiología , Enfermedad de Paget Extramamaria/etiología , Enfermedad de Paget Extramamaria/microbiología , Staphylococcus aureus/aislamiento & purificación , Anciano , Anciano de 80 o más Años , Correlación de Datos , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA