Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Clin Endocrinol Metab ; 103(11): 4241-4252, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30124866

RESUMEN

Context: Fos null mice failed to ovulate and form a corpus luteum (CL) even when given exogenous gonadotropins, suggesting that ovarian Fos expression is critical for successful ovulation and CL formation. However, little is known about FOS in the human ovary. Objectives: To determine the expression, regulation, and function of FOS in human periovulatory follicles. Design/Participants: Timed periovulatory follicles were obtained from normally cycling women. Granulosa/lutein cells were collected from in vitro fertilization patients. Main Outcome Measures: The in vivo expression after human chorionic gonadotropin (hCG) administration and in vitro regulation of FOS, JUN, JUNB, and JUND was evaluated at the mRNA and protein level. Binding of progesterone receptor (PGR) and FOS to their target genes was assessed by chromatin immunoprecipitation analyses. Prostaglandin E2 (PGE2) and progesterone were measured. Results: The expression of FOS, JUNB, and JUND drastically increased in ovulatory follicles after hCG administration. In human granulosa/lutein cell cultures, hCG increased the expression of FOS and JUN proteins. Inhibitors of PGR and epidermal growth factor (EGF) receptors reduced hCG-induced increases in the expression and phosphorylation of FOS. PGR bound to the FOS gene. A selective FOS inhibitor blocked hCG-induced increases in PGE2 and the expression of prostaglandin (PG) synthases and transporters (PTGES, SLCO2A1, and ABCC1). FOS bound to the promoter regions of these genes. Conclusions: The increase of FOS/activator protein 1 in human periovulatory follicles after hCG administration is mediated by collaborative actions of PGR and EGF signaling and critical for the upregulated expression of key ovulatory genes required for the rise in ovulatory PG in human granulosa cells.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Folículo Ovárico/metabolismo , Ovulación/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Adulto , Benzofenonas/farmacología , Células Cultivadas , Dinoprostona/análisis , Dinoprostona/metabolismo , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Humanos , Isoxazoles/farmacología , Mifepristona/farmacología , Folículo Ovárico/citología , Cultivo Primario de Células , Progesterona/análisis , Progesterona/metabolismo , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fos/genética , Quinazolinas/farmacología , ARN Interferente Pequeño/metabolismo , Receptores de Progesterona/antagonistas & inhibidores , Receptores de Progesterona/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Tirfostinos/farmacología , Regulación hacia Arriba
2.
Endocrinology ; 159(6): 2447-2458, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29648638

RESUMEN

The luteinizing hormone (LH) surge is essential for ovulation, but the intrafollicular factors induced by LH that mediate ovulatory processes (e.g., angiogenesis) are poorly understood, especially in women. The role of secretogranin II (SCG2) and its cleaved bioactive peptide, secretoneurin (SN), were investigated as potential mediators of ovulation by testing the hypothesis that SCG2/SN is induced in granulosa cells by human chorionic gonadotropin (hCG), via a downstream LH receptor signaling mechanism, and stimulates ovarian angiogenesis. Humans, nonhuman primates, and rodents were treated with hCG in vivo resulting in a significant increase in the messenger RNA and protein levels of SCG2 in granulosa cells collected early during the periovulatory period and just prior to ovulation (humans: 12 to 34 hours; monkeys: 12 to 36 hours; rodents: 4 to 12 hours post-hCG). This induction by hCG was recapitulated in an in vitro culture system utilizing granulosa-lutein cells from in vitro fertilization patients. Using this system, inhibition of downstream LH receptor signaling pathways revealed that the initial induction of SCG2 is regulated, in part, by epidermal growth factor receptor signaling. Further, human ovarian microvascular endothelial cells were treated with SN (1 to 100 ng/mL) and subjected to angiogenesis assays. SN significantly increased endothelial cell migration and new sprout formation, suggesting induction of ovarian angiogenesis. These results establish that SCG2 is increased in granulosa cells across species during the periovulatory period and that SN may mediate ovulatory angiogenesis in the human ovary. These findings provide insight into the regulation of human ovulation and fertility.


Asunto(s)
Células de la Granulosa/metabolismo , Neovascularización Fisiológica/genética , Ovario/irrigación sanguínea , Ovulación/genética , Secretogranina II/genética , Adulto , Animales , Células Cultivadas , Femenino , Humanos , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Ovario/metabolismo , Ratas , Ratas Sprague-Dawley , Secretogranina II/metabolismo , Regulación hacia Arriba/genética
3.
Biol Reprod ; 96(6): 1256-1266, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28595291

RESUMEN

The chemokine CXC motif ligand 12 (CXCL12) and its cognate receptor, CXCR4, have been implicated in the ovulatory process in various animal models. However, little is known about the expression and regulation of CXCL12 and CXCR4 and their functions during the ovulatory period in the human ovary. In this study, we characterized the expression patterns of CXCL12 and CXCR4 in preovulatory follicles collected before the luteinizing hormone (LH) surge and at defined hours after hCG administration in women with the regular menstrual cycle. The levels of mRNA and protein for CXCR4 were increased in granulosa cells of late ovulatory follicles, whereas CXCL12 expression was constant in follicles throughout the ovulatory period. Both CXCR4 and CXCL12 were localized to a subset of leukocytes around and inside the vasculature of human preovulatory follicles. Using a human granulosa cell culture model, the regulatory mechanisms and functions of CXCL12 and CXCR4 expression were investigated. Human chorionic gonadotropin (hCG) stimulated CXCR4 expression, whereas CXCL12 expression was not affected, mimicking in vivo expression patterns. Both RU486 (progesterone receptor antagonist) and CoCl2 (HIFs activator) blocked the hCG-induced increase in CXCR4 expression, whereas AG1478 (EGFR inhibitor) had no effect. The treatment with CXCL12 had no effect on granulosa cell viability but decreased hCG-stimulated CXCR4 expression.


Asunto(s)
Células de la Granulosa/fisiología , Compuestos Heterocíclicos/farmacología , Hormona Luteinizante/metabolismo , Ovulación/fisiología , Receptores CXCR4/metabolismo , Receptores de Progesterona/fisiología , Bencilaminas , Supervivencia Celular , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacología , Gonadotropina Coriónica/farmacología , Ciclamas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR4/genética , Técnicas de Cultivo de Tejidos
4.
J Clin Endocrinol Metab ; 102(6): 1971-1982, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28323945

RESUMEN

Context: In animal models, the luteinizing hormone surge increases progesterone (P4) and progesterone receptor (PGR), prostaglandins (PTGs), and epidermal growth factor (EGF)-like factors that play essential roles in ovulation. However, little is known about the expression, regulation, and function of these key ovulatory mediators in humans. Objective: To determine when and how these key ovulatory mediators are induced after the luteinizing hormone surge in human ovaries. Design and Participants: Timed periovulatory follicles were obtained from cycling women. Granulosa/lutein cells were collected from in vitro fertilization patients. Main Outcome Measures: The in vivo and in vitro expression of PGR, PTG synthases and transporters, and EGF-like factors were examined at the level of messenger RNA and protein. PGR binding to specific genes was assessed. P4 and PTGs in conditioned media were measured. Results: PGR, PTGS2, and AREG expressions dramatically increased in ovulatory follicles at 12 to 18 hours after human chorionic gonadotropin (hCG). In human granulosa/lutein cell cultures, hCG increased P4 and PTG production and the expression of PGR, specific PTG synthases and transporters, and EGF-like factors, mimicking in vivo expression patterns. Inhibitors for P4/PGR and EGF-signaling pathways reduced hCG-induced increases in PTG production and the expression of EGF-like factors. PGR bound to the PTGS2, PTGES, and SLCO2A1 genes. Conclusions: This report demonstrated the time-dependent induction of PGR, AREG, and PTGS2 in human periovulatory follicles. In vitro studies indicated that collaborative actions of P4/PGR and EGF signaling are required for hCG-induced increases in PTG production and potentiation of EGF signaling in human periovulatory granulosa cells.


Asunto(s)
Anfirregulina/genética , Ciclooxigenasa 2/genética , Transportadores de Anión Orgánico/genética , Folículo Ovárico/metabolismo , Progesterona/metabolismo , Prostaglandinas/metabolismo , Receptores de Progesterona/genética , Adulto , Anfirregulina/metabolismo , Western Blotting , Células Cultivadas , Gonadotropina Coriónica/farmacología , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Fertilización In Vitro , Perfilación de la Expresión Génica , Células de la Granulosa , Humanos , Inmunohistoquímica , Células Lúteas , Hormona Luteinizante , Transportadores de Anión Orgánico/efectos de los fármacos , Transportadores de Anión Orgánico/metabolismo , Ovulación , Reacción en Cadena de la Polimerasa , Prostaglandina-E Sintasas/efectos de los fármacos , Prostaglandina-E Sintasas/genética , Prostaglandina-E Sintasas/metabolismo , ARN Mensajero/metabolismo , Receptores de Progesterona/efectos de los fármacos , Receptores de Progesterona/metabolismo
5.
Endocrinology ; 158(1): 109-120, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27813674

RESUMEN

Increased proteolytic activity is a key event that aids in breakdown of the follicular wall to permit oocyte release. How the protease activity is regulated is still unknown. We hypothesize that tissue factor pathway inhibitor 2 (TFPI2), a Kunitz-type serine protease inhibitor, plays a role in regulating periovulatory proteolytic activity as in other tissues. TFPI2 is secreted into the extracellular matrix (ECM) where it is postulated to regulate physiological ECM remodeling. The expression profile of TFPI2 during the periovulatory period was assessed utilizing a well-characterized human menstrual cycle model and a gonadotropin-primed rat model. Administration of an ovulatory dose of human chorionic gonadotropin (hCG) increased TFPI2 expression dramatically in human and rat granulosa and theca cells. This increase in Tfpi2 expression in rat granulosa cells required hCG-mediated epidermal growth factor, protein kinase A, mitogen-activated protein kinase (MAPK) 1/2, p38 MAPK and protease activated receptor 1-dependent cell signaling. A small interferingRNA-mediated knockdown of TFPI2 in rat granulosa cells resulted in increased plasmin activity in the granulosa cell conditioned media. Knockdown of TFPI2 also reduced expression of multiple genes including interleukin 6 (Il6) and amphiregulin (Areg). Overexpression of TFPI2 using an adenoviral vector partially restored the expression of Il6 and Areg in TFPI2 siRNA treated rat granulosa cells. These data support the hypothesis that TFPI2 is important for moderating plasmin activity and regulating granulosa cell gene expression during the periovulatory period. We, therefore, propose that through these actions, TFPI2 aids in the tissue remodeling taking place during follicular rupture and corpus luteum formation.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Fibrinolisina/metabolismo , Glicoproteínas/metabolismo , Células de la Granulosa/metabolismo , Ovulación , Animales , Células Cultivadas , Femenino , Humanos , Ratas Sprague-Dawley , Células Tecales/metabolismo
6.
Fertil Steril ; 103(3): 826-33, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25516084

RESUMEN

OBJECTIVE: To explore the temporal expression in granulosa and theca cells of key members of the MMP and ADAMTS families across the periovulatory period in women to gain insight into their possible roles during ovulation and early luteinization. DESIGN: Experimental prospective clinical study and laboratory-based investigation. SETTING: University medical center and private IVF center. ANIMAL AND PATIENT(S): Thirty-eight premenopausal women undergoing surgery for tubal ligation and six premenopausal women undergoing assisted reproductive techniques. INTERVENTION(S): Administration of hCG and harvesting of follicles by laparoscopy and collection of granulosa-lutein cells at oocyte retrieval. MAIN OUTCOME MEASURE(S): Expression of mRNA for matrix metalloproteinase (MMPs) and the A disintegrin and metalloproteinase with thrombospondin-like motifs (ADAMTS) in human granulosa cells and theca cells collected across the periovulatory period of the menstrual cycle and in cultured granulosa-lutein cells after hCG. Localization of MMPs and ADAMTSs by immunohistochemistry. RESULT(S): Expression of MMP1 and MMP19 mRNA increased in both granulosa and theca cells after hCG administration. ADAMTS1 and ADAMTS9 mRNA increased in granulosa cells after hCG treatment, however, thecal cell expression for ADAMTS1 was unchanged, while ADAMTS9 expression was decreased. Expression of MMP8 and MMP13 mRNA was unchanged. Immunohistochemistry confirmed the localization of MMP1, MMP19, ADAMTS1, and ADAMTS9 to the granulosa and thecal cell layers. CONCLUSION(S): The collection of the dominant follicle throughout the periovulatory period has allowed the identification of proteolytic remodeling enzymes in the granulosa and theca compartments that may be critically involved in human ovulation. These proteinases may work in concert to regulate breakdown of the follicular wall and release of the oocyte.


Asunto(s)
Gonadotropina Coriónica/farmacología , Fase Folicular , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Péptido Hidrolasas/biosíntesis , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Células Cultivadas , Inducción Enzimática/efectos de los fármacos , Femenino , Fase Folicular/efectos de los fármacos , Fase Folicular/genética , Fase Folicular/metabolismo , Humanos , Células Lúteas/efectos de los fármacos , Células Lúteas/metabolismo , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Folículo Ovárico/enzimología
7.
Biol Reprod ; 91(2): 34, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24920038

RESUMEN

An intrafollicular increase in proteolytic activity drives ovulatory events. Surprisingly, the periovulatory expression profile of the membrane-type matrix metalloproteinases (MT-MMPs), unique proteases anchored to the cell surface, has not been extensively examined. Expression profiles of the MT-MMPs were investigated in ovarian tissue from well-characterized rat and macaque periovulatory models and naturally cycling women across the periovulatory period. Among the six known MT-MMPs, mRNA expression of Mmp14, Mmp16, and Mmp25 was increased after human chorionic gonadotropin (hCG) administration in rats. In human granulosa cells, mRNA expression of MMP14 and MMP16 increased following hCG treatment. In contrast, mRNA levels of MMP16 and MMP25 in human theca cells were unchanged before ovulation but declined by the postovulatory stage. In macaque granulosa cells, hCG increased mRNA for MMP16 but not MMP14. Immunoblotting showed that protein levels of MMP14 and MMP16 in rats increased, similar to their mRNA expression. In macaque granulosa cells, only the active form of the MMP14 protein increased after hCG, unlike its mRNA or the proprotein. By immunohistochemistry, both MMP14 and MMP16 localized to the different ovarian cell types in rats and humans. Treatment with hCG resulted in intense immunoreactivity of MMP14 and MMP16 proteins in the granulosa and theca cells. The present study shows that MMP14 and MMP16 are increased by hCG administration in the ovulating follicle, demonstrating that these MMPs are conserved among rats, macaques, and humans. These findings suggest that MT-MMPs could have an important role in promoting ovulation and remodeling of the ovulated follicle into the corpus luteum.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Macaca fascicularis/fisiología , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 16 de la Matriz/metabolismo , Ovario/enzimología , Ovulación/fisiología , Animales , Femenino , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Humanos , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 16 de la Matriz/genética , Metaloproteinasas de la Matriz Asociadas a la Membrana/genética , Metaloproteinasas de la Matriz Asociadas a la Membrana/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Transcriptoma
8.
Biol Reprod ; 89(5): 121, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24048576

RESUMEN

Ovulation involves reorganization of the extracellular matrix of the follicle. This study examines the expression, localization, and potential function of the tissue inhibitor of metalloproteinase 3 (TIMP3) during ovulation in women. The dominant follicle of the menstrual cycle was collected at specified times throughout the ovulatory process: pre-, early, late, and postovulatory. For quantitative studies, the follicle was bisected; granulosa and theca cells were separated and collected. For immunohistochemistry (IHC), the intact follicle was embedded and TIMP3 was localized. Additionally, granulosa cells were collected from women undergoing in vitro fertilization and treated with increasing concentrations of recombinant TIMP3, and cell viability was assessed. Real-time PCR for TIMP3 mRNA revealed an increase in TIMP3 mRNA expression in granulosa cells from the early to the late ovulatory stage. Thecal TIMP3 mRNA expression was constitutive across the periovulatory period. TIMP3 protein was localized by IHC to the granulosa and theca cell layers in pre-, early, and late ovulatory follicles as well as to the vascular bed. The staining was most intense in the granulosa and theca cells in the late ovulatory group. Treatment of human granulosa-lutein cells with exogenous recombinant TIMP3 for 24 h decreased cell viability by 60%. Using human follicles collected throughout the periovulatory period of the menstrual cycle, we have demonstrated that TIMP3 mRNA expression increases and that TIMP3 protein is in the appropriate cellular layers to regulate proteolytic remodeling as the follicle progresses toward ovulation. In addition, we have shown that elevated levels of TIMP3 lead to decreased cell viability.


Asunto(s)
Ovario/enzimología , Ovulación/metabolismo , Inhibidor Tisular de Metaloproteinasa-3/genética , Inhibidor Tisular de Metaloproteinasa-3/metabolismo , Adulto , Células Cultivadas , Gonadotropina Coriónica/farmacología , Femenino , Expresión Génica , Humanos , Células Lúteas/efectos de los fármacos , Células Lúteas/fisiología , Ciclo Menstrual/efectos de los fármacos , Ciclo Menstrual/genética , Ciclo Menstrual/metabolismo , Recuperación del Oocito , Ovulación/genética , Distribución Tisular , Inhibidor Tisular de Metaloproteinasa-3/farmacología
9.
Biol Reprod ; 86(3): 78, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22116802

RESUMEN

The matrix metalloproteinases (MMPs) are postulated to facilitate follicular rupture. In the present study, expression of the stromelysins (MMP3, MMP10, MMP11) was analyzed in the periovulatory human and rat ovary. Human granulosa and theca cells were collected from the dominant follicle at various times after human chorionic gonadotropin (hCG). Intact rat ovaries, granulosa cells, and residual tissue (tissue remaining after granulosa cell collection) were isolated from equine CG (eCG)-hCG-primed animals. Mmp10 mRNA was highly induced in human granulosa and theca cells and intact rat ovaries, granulosa cells, and residual tissue. Localization of MMP10 to granulosa and theca cells in both human and rat ovarian follicles was confirmed by immunohistochemistry. Mmp3 mRNA was unchanged in human cells and rat granulosa cells, but increased in intact rat ovaries and residual tissue. Mmp11 mRNA decreased following hCG treatment in human granulosa and theca cells as well as rat granulosa cells. Regulation of Mmp10 in cultured rat granulosa cells revealed that the EGF inhibitor AG1478 and the progesterone receptor antagonist RU486 suppressed the induction of Mmp10 mRNA, whereas the prostaglandin inhibitor NS398 had no effect. Studies on the Mmp10 promoter demonstrated that forskolin plus PMA stimulated promoter activity, which was dependent upon a proximal AP1 site. In conclusion, there are divergent patterns of stromelysin expression associated with ovulation, with a marked induction of Mmp10 mRNA and a decrease in Mmp11 mRNA, yet a species-dependent pattern on Mmp3 mRNA expression. The induction of Mmp10 expression suggests an important role for this MMP in the follicular changes associated with ovulation and subsequent luteinization.


Asunto(s)
Células de la Granulosa/metabolismo , Metaloproteinasa 10 de la Matriz/metabolismo , Metaloproteinasa 11 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Ovario/metabolismo , Ovulación/metabolismo , Células Tecales/metabolismo , Animales , Células Cultivadas , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Femenino , Células de la Granulosa/citología , Células de la Granulosa/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Humanos , Mifepristona/farmacología , Ovario/citología , Ovario/efectos de los fármacos , Quinazolinas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Células Tecales/citología , Células Tecales/efectos de los fármacos , Tirfostinos/farmacología
10.
Methods Mol Biol ; 590: 115-29, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19763500

RESUMEN

In situ hybridization represents a powerful technique to localize DNA or RNA of interest at the chromosomal or cellular level. In endocrine tissues composed of diverse and varied cell types, in situ hybridization has allowed the identification of specific cells responsible for the expression of genes controlling the function of the tissue. Our laboratory has routinely used this approach to understand the cellular expression of genes associated with the growth of the ovarian follicle, rupture of the follicle, and transformation of the ruptured follicle into the corpus luteum. The current study outlines the procedural details of in situ detection of mRNA in tissues and illustrates the utility of this approach in identifying the ovarian cells expressing the matrix metalloproteinases and their endogenous inhibitors, the TIMPs, in the human ovary.


Asunto(s)
Metaloproteinasas de la Matriz/genética , Ovario/enzimología , ARN Mensajero/genética , Femenino , Humanos , Hibridación in Situ
11.
J Neurosci ; 25(1): 62-7, 2005 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-15634767

RESUMEN

Input from the suprachiasmatic nucleus (SCN) to gonadotropin-releasing hormone (GnRH) neurons is critical to the occurrence of regular cyclic GnRH secretion. It is thought that an essential neuropeptide in the SCN that communicates this cyclic information to GnRH neurons is vasoactive intestinal polypeptide (VIP) and that it may act through cAMP. We tested the hypothesis that (1) aging involves a blunting of cAMP diurnal rhythmicity in the SCN; (2) administration of antisense oligonucleotides (anti-oligos) against VIP, which produces an aging-like pattern in VIP, would lead to an aging-like suppression of cAMP; and (3) this in turn would lead to inhibition of the steroid-induced activation of GnRH neurons. We measured cAMP concentrations in the SCN and rostral preoptic nucleus throughout the day in young and middle-aged rats that were ovariectomized (OVX) or OVX and treated with estradiol. Our results show that cAMP concentrations exhibit a diurnal rhythm in young rats, and that this rhythm is totally abolished by the time rats are middle age. Administration of antisense oligonucleotides against VIP or random oligos suppresses VIP concentrations and abolishes the cAMP rhythm, leading to significantly reduced activation of GnRH neurons. Together, these findings strongly suggest that the SCN conveys diurnal information to GnRH neurons by driving VIP-dependent cAMP rhythms. In addition, aging involves deterioration in this VIP-driven rhythmicity, which impacts the ability of steroids to induce GnRH neuronal activation.


Asunto(s)
Envejecimiento/metabolismo , Ritmo Circadiano/fisiología , AMP Cíclico/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Núcleo Supraquiasmático/fisiología , Péptido Intestinal Vasoactivo/metabolismo , Animales , Corteza Cerebral/metabolismo , Estradiol/farmacología , Femenino , Hipotálamo/metabolismo , Núcleos Talámicos de la Línea Media/metabolismo , Neuronas/metabolismo , Oligodesoxirribonucleótidos Antisentido , Ovariectomía , Área Preóptica/metabolismo , Ratas , Ratas Sprague-Dawley , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/metabolismo , Péptido Intestinal Vasoactivo/antagonistas & inhibidores
12.
Brain Res ; 990(1-2): 87-94, 2003 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-14568333

RESUMEN

Estrogen-related changes in serotonergic neuronal transmission, including changes in the number of serotonin transporter (SERT) binding sites, have been cited as a possible cause for changes in mood, memory and sleep that occur during the menopausal transition. However, both aging and estradiol regulate SERT binding sites in the brain. The goal of this experiment was to determine how aging and estrogen interact to regulate SERT levels in the forebrain of young and reproductively senescent female Sprague-Dawley rats using [3H]paroxetine. The density of specific [3H]paroxetine binding in various brain regions was compared in young (2-4 months) and reproductively senescent (10-12 months) female rats at three times of day. In most brain regions examined, estrogen and aging independently increased the number of [3H]paroxetine binding sites. The only region that displayed a reduction in [3H]paroxetine binding with age was the suprachiasmatic nucleus (SCN). Time of day influenced [3H]paroxetine binding in the SCN and the paraventricular thalamus (PVT), two regions known to be involved in the regulation of circadian rhythms. Aging and/or estrogen also altered the pattern of binding in these regions. Thus, based on the results of this study, we conclude that aging and estrogen both act to regulate SERT binding sites in the forebrain of female rats, and that this regulation is region specific.


Asunto(s)
Envejecimiento/metabolismo , Proteínas Portadoras/metabolismo , Sistema Nervioso Central/metabolismo , Ritmo Circadiano/fisiología , Estradiol/farmacología , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana , Proteínas del Tejido Nervioso , Animales , Autorradiografía , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/fisiología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ovariectomía , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Paroxetina/metabolismo , Paroxetina/farmacología , Unión Proteica , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Inhibidores Selectivos de la Recaptación de Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Sueño/fisiología , Núcleo Supraquiasmático/efectos de los fármacos , Núcleo Supraquiasmático/metabolismo , Vigilia/fisiología
13.
Recent Prog Horm Res ; 57: 235-56, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12017546

RESUMEN

The menopause marks the end of a woman's reproductive life. During the postmenopausal period, plasma estrogen concentrations decrease dramatically and remain low for the rest of her life, unless she chooses to take hormone replacement therapy. During the past 20 years, we have learned that changes in the central nervous system are associated with and may influence the timing of the menopause in women. Recently, it has become clear that estrogens act on more than just the hypothalamus, pituitary, ovary, and other reproductive organs. In fact, they play roles in a wide variety of nonreproductive functions. With the increasing life span of humans from approximately 50 to 80 years and the relatively fixed age of the menopause, a larger number of women will spend over one third of their lives in the postmenopausal state. It is not surprising that interest has increased in factors that govern the timing of the menopause and the repercussions of the lack of estrogen on multiple aspects of women's health. We have used animal models to better understand the complex interactions between the ovary and the brain that lead to the menopause and the repercussions of the hypoestrogenic state. Our results show that when rats reach middle age, the patterns and synchrony of multiple neurochemical events that are critical to the preovulatory gonadotropin-releasing hormone (GnRH) surge undergo subtle changes. The precision of rhythmic pattern of neurotransmitter dynamics depends on the presence of estradiol. Responsiveness to this hormone decreases in middle-aged rats. The lack of precision in the coordination in the output of neural signals leads to a delay and attenuation of the luteinizing hormone surge, which lead to irregular estrous cyclicity and, ultimately, to the cessation of reproductive cycles. We also have examined the impact of the lack of estrogen on the vulnerability of the brain to injury. Our work establishes that the absence of estradiol increases the extent of cell death after stroke-like injury and that treatment with low physiological levels of estradiol are profoundly neuroprotective. We have begun to explore the cellular and molecular mechanisms that underlie this novel nonreproductive action of estrogens. In summary, our studies show that age-related changes in the ability of estradiol to coordinate the neuroendocrine events that lead to regular preovulatory GnRH surges contribute to the onset of irregular estrous cycles and eventually to acyclicity. Furthermore, we have shown that the lack of estradiol increases the vulnerability of the brain to injury and neurodegeneration.


Asunto(s)
Envejecimiento , Menopausia , Sistemas Neurosecretores/fisiología , Reproducción , Animales , Encéfalo/fisiología , Estradiol/fisiología , Femenino , Hormona Liberadora de Gonadotropina/fisiología , Gonadotropinas Hipofisarias/metabolismo , Humanos , Persona de Mediana Edad , Ovario/fisiología
14.
Mech Ageing Dev ; 123(6): 593-601, 2002 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-11850023

RESUMEN

Estradiol's ability to influence neurochemical events that are critical to female reproductive cyclicity and behavior decreases with age. We tested the hypothesis that decreases in estrogen receptor-alpha (ERalpha) and/or ERbeta mRNA explain the brain's declining responsiveness to estradiol. We assessed ERalpha and ERbeta mRNA levels in intact and ovariectomized estradiol-treated rats. ERbeta mRNA was detected in several brain regions and decreased by middle-age in the cerebral cortex and supraoptic nucleus of estradiol-treated rats. ERbeta mRNA levels exhibited a diurnal rhythm in the suprachiasmatic nucleus of young and middle-aged rats and this rhythm was blunted in old rats. We examined ERalpha mRNA in the periventricular preoptic, medial preoptic, ventromedial and arcuate nuclei, and it was decreased only in the periventricular preoptic nucleus of the old rats. In summary, the expression of ERalpha and ERbeta mRNAs is differentially modulated in the aging brain and changes are region specific.


Asunto(s)
Envejecimiento/genética , Encéfalo/metabolismo , Expresión Génica , Receptores de Estrógenos/genética , Envejecimiento/metabolismo , Animales , Corteza Cerebral/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Femenino , Ovariectomía , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Núcleo Supraquiasmático/metabolismo , Núcleo Supraóptico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...