Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroendocrinol ; 31(12): e12809, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31715031

RESUMEN

An injection of unesterified oestradiol (E2 ) facilitates receptive behaviour in E2 benzoate (EB)-primed, ovariectomised female rats when it is administered i.c.v. or systemically. The present study tested the hypothesis that inhibitors of protein kinase A (PKA), protein kinase G (PKG) or the Src/mitogen-activated protein kinase (MAPK) complex interfere with E2 facilitation of receptive behaviour. In Experiment 1, lordosis induced by i.c.v. infusion of E2 was significantly reduced by i.c.v. administration of Rp-cAMPS, a PKA inhibitor, KT5823, a PKG inhibitor, and PP2 and PD98059, Src and MAPK inhibitors, respectively, between 30 and 240 minutes after infusion. In Experiment 2, we determined whether the ventromedial hypothalamus (VMH) is one of the neural sites at which those intracellular pathways participate in lordosis behaviour induced by E2 . Administration of each of the four protein kinase inhibitors into the VMH blocked facilitation of lordosis induced by infusion of E2 also into the VMH. These data support the hypothesis that activation of several protein kinase pathways is involved in the facilitation of lordosis by E2 in EB-primed rats.


Asunto(s)
Antagonistas de Estrógenos/farmacología , Lordosis/fisiopatología , Inhibidores de Proteínas Quinasas/farmacología , Núcleo Hipotalámico Ventromedial/fisiología , Animales , Carbazoles/farmacología , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Estradiol/fisiología , Femenino , Flavonoides/farmacología , Infusiones Intraventriculares , Lordosis/inducido químicamente , Masculino , Microinyecciones , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirimidinas/farmacología , Ratas , Tionucleótidos/farmacología , Núcleo Hipotalámico Ventromedial/efectos de los fármacos
2.
Horm Behav ; 86: 1-7, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27594441

RESUMEN

The present study was designed to assess the participation of estrogen receptors alpha (ERα) and beta (ERß) in the short-term facilitation of lordosis behavior in ovariectomized (ovx), estradiol (E2) primed rats. In experiment 1, dose response curves for PPT and DPN (ERα and ERß agonists, respectively) facilitation of lordosis behavior (lordosis quotient and lordosis score) were established by infusing these agonists into the right lateral ventricle (icv) in female rats injected 40h previously with 5µg of E2 benzoate. PPT doses of 0.08 and 0.4ng produced high lordosis quotients starting at 30min and continuing at 120 and 240min post-injection. DPN induced high levels of lordosis behavior at all times tested. However, the intensity of lordosis induced by both agonists was weak. In experiment 2, we tested the involvement of each ER in facilitation of lordosis by icv infusion of MPP (ERα-selective antagonist) or PHTPP (ERß-selective antagonist) prior to infusion of 2ng of free E2. Icv infusion of either MPP or PHTPP 30min before free E2 significantly depressed E2 facilitation of lordosis. The results suggest that both forms of ER are involved in the short-latency facilitation of lordosis behavior in E2-primed rats.


Asunto(s)
Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/fisiología , Postura/fisiología , Conducta Sexual Animal/fisiología , Animales , Estradiol/farmacología , Receptor alfa de Estrógeno/agonistas , Receptor beta de Estrógeno/agonistas , Ciclo Estral/efectos de los fármacos , Ciclo Estral/fisiología , Femenino , Masculino , Ovariectomía , Ratas , Ratas Sprague-Dawley , Conducta Sexual Animal/efectos de los fármacos
3.
Endocrinology ; 155(5): 1827-37, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24617524

RESUMEN

This study investigated potential mechanisms by which age and IGF-I receptor (IGF-Ir) signaling in the neuroendocrine hypothalamus affect estradiol-positive feedback effects on GnRH neuronal activation and on kisspeptin and N-methyl-D-aspartate (NMDA)-induced LH release and on the abundance of NMDA receptor subunits Nr1 and Nr2b and Kiss1r transcript and protein in the hypothalamus of young and middle-aged female rats. We infused vehicle, IGF-I, or JB-1, a selective antagonist of IGF-Ir, into the third ventricle of ovariectomized female rats primed with estradiol or vehicle and injected with vehicle, kisspeptin (3 or 30 nmol/kg), or NMDA (15 or 30 mg/kg). Regardless of dose, NMDA and kisspeptin resulted in significantly more LH release, GnRH/c-Fos colabeling, and c-Fos immunoreative cells in young than in middle-aged females. Estradiol priming significantly increased Kiss1r, Nr1, and Nr2b receptor transcript and protein abundance in young but not middle-aged female hypothalamus. JB-1 attenuated kisspeptin and NMDA-induced LH release, numbers of GnRH/c-Fos and c-Fos cells, and Kiss1r, Nr1, and Nr2b transcript and protein abundance in young females to levels observed in middle-aged females. IGF-I significantly enhanced NMDA and kisspeptin-induced LH release in middle-aged females without increasing numbers of GnRH/c-Fos or c-Fos immunoreactive cells. IGF-I infusion in middle-aged females also increased Kiss1r, Nr1, and Nr2b protein and transcript to levels that were equivalent to young estradiol-primed females. These findings indicate that age-related changes in estradiol-regulated responsiveness to excitatory input from glutamate and kisspeptin reflect reduced IGF-Ir signaling.


Asunto(s)
Envejecimiento , Factor I del Crecimiento Similar a la Insulina/metabolismo , Kisspeptinas/metabolismo , Hormona Luteinizante/metabolismo , Receptor IGF Tipo 1/agonistas , Receptores de N-Metil-D-Aspartato/agonistas , Transmisión Sináptica , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Infusiones Intraventriculares , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/análogos & derivados , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , N-Metilaspartato/metabolismo , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células Neuroendocrinas/citología , Células Neuroendocrinas/efectos de los fármacos , Células Neuroendocrinas/metabolismo , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos
4.
J Neurosci ; 33(30): 12364-74, 2013 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-23884942

RESUMEN

Transient global ischemia causes selective, delayed death of hippocampal CA1 pyramidal neurons in humans and animals. It is well established that estrogens ameliorate neuronal death in animal models of focal and global ischemia. However, the role of signal transducer and activator of transcription-3 (STAT3) and its target genes in estradiol neuroprotection in global ischemia remains unclear. Here we show that a single intracerebral injection of 17ß-estradiol to ovariectomized female rats immediately after ischemia rescues CA1 neurons destined to die. Ischemia promotes activation of STAT3 signaling, association of STAT3 with the promoters of target genes, and STAT3-dependent mRNA and protein expression of prosurvival proteins in the selectively vulnerable CA1. In animals subjected to ischemia, acute postischemic estradiol further enhances activation and nuclear translocation of STAT3 and STAT3-dependent transcription of target genes. Importantly, we show that STAT3 is critical to estradiol neuroprotection, as evidenced by the ability of STAT3 inhibitor peptide and STAT3 shRNA delivered directly into the CA1 of living animals to abolish neuroprotection. In addition, we identify survivin, a member of the inhibitor-of-apoptosis family of proteins and known gene target of STAT3, as essential to estradiol neuroprotection, as evidenced by the ability of shRNA to survivin to reverse neuroprotection. These findings indicate that ischemia and estradiol act synergistically to promote activation of STAT3 and STAT3-dependent transcription of survivin in insulted CA1 neurons and identify STAT3 and survivin as potentially important therapeutic targets in an in vivo model of global ischemia.


Asunto(s)
Isquemia Encefálica/fisiopatología , Estradiol/fisiología , Proteínas Asociadas a Microtúbulos/genética , Factor de Transcripción STAT3/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Isquemia Encefálica/tratamiento farmacológico , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Estradiol/farmacología , Femenino , Inyecciones Intraventriculares , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fármacos Neuroprotectores/farmacología , Ovariectomía , Fosforilación/fisiología , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/genética , Survivin
5.
Pharmacol Biochem Behav ; 110: 13-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23743347

RESUMEN

The present study tested the hypothesis that the Janus kinase 2, Src tyrosine kinases, and mitogen-activated protein kinase interact to regulate lordosis behavior induced by leptin in ovariectomized, estrogen-primed rats. The role of protein kinase A and protein kinase C in lordosis facilitation by leptin was also assessed. In experiment 1, the intracerebroventricular administration of leptin to ovariectomized, estradiol-primed rats significantly stimulated lordosis behavior at 1, 2 and 4 h post-injection tests. In experiment 2, the Janus kinase 2 inhibitor AG490, the Src tyrosine kinase inhibitor PP2 and the mitogen-activated protein kinase inhibitor PD98059 were administered into the right lateral ventricle before leptin. The lordosis quotient and the lordosis score induced by leptin were significantly decreased by each of these kinase inhibitors. In experiment 3, we examined the effects of RpcAMPS and bisindolylmaleimide, protein kinase A and protein kinase C inhibitors on the lordosis elicited by leptin administration. Lordosis behavior induced by leptin was significantly decreased by both the protein kinase A and protein kinase C inhibitors at 1 h post-leptin injection. The results confirm that multiple intracellular pathways participate in the expression of lordosis behavior in estrogen-primed rats elicited by leptin.


Asunto(s)
Dorso/fisiología , Estrógenos/administración & dosificación , Leptina/fisiología , Ovariectomía , Proteínas Quinasas/metabolismo , Conducta Sexual Animal , Animales , Femenino , Infusiones Intraventriculares , Leptina/administración & dosificación , Ratas , Ratas Sprague-Dawley
6.
Steroids ; 78(6): 597-606, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23385013

RESUMEN

The ovarian hormone 17ß-estradiol (E2) exerts profound neuroprotective actions against ischemia-induced brain damage in rodent models of global and focal ischemia. This review focuses on the neuroprotective efficacy of post-ischemic administration of E2 and non-feminizing estrogen analogs in the aging brain, with an emphasis on studies in animals subjected to a long-term loss of circulating E2. Clinical findings from the Women's Health Initiative study as well as data from animal studies that used long-term, physiological levels of E2 treatment are discussed in this context. We summarize major published findings that highlight the effective doses and timing of E2 treatment relative to onset of ischemia. We then discuss recent findings from our laboratory showing that under some conditions the aging hippocampus remains responsive to E2 and some neuroprotective non-feminizing estrogen analogs even after prolonged periods of hormone withdrawal. Possible membrane-initiated signaling mechanisms that may underlie the neuroprotective actions of acutely administered E2 are also discussed. Based on these findings, we suggest that post-ischemic treatment with high doses of E2 or certain non-feminizing estrogen analogs may have great therapeutic potential for treatment of brain damage and neurodegeneration associated with ischemia.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Modelos Animales de Enfermedad , Estradiol/uso terapéutico , Estrógenos/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Animales , Relación Dosis-Respuesta a Droga , Estradiol/administración & dosificación , Estrógenos/administración & dosificación , Humanos , Fármacos Neuroprotectores/administración & dosificación
7.
Horm Behav ; 62(5): 579-84, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23010621

RESUMEN

The progesterone receptor (PR) is a dual function protein that acts in the nucleus as a transcriptional factor and at the cytoplasm as a scaffold for the Src-MAPK signaling pathway. Several agents lacking affinity for the PR, such as 5ß-reduced progestins, GnRH or prostaglandin E(2) (PGE(2)) facilitate estrous behavior in ovariectomized (ovx), estrogen-primed rats yet their action is blocked by the antiprogestin RU486. We hypothesize that these agents act by using the PR-Src-mitogen activated protein kinase alternative pathway. To test this hypothesis we used PP2, a specific inhibitor of the Src kinase family. Intraventricular infusion of 30 µg of PP2, 30 min before behavioral testing, significantly attenuated estrous behaviors induced in estradiol benzoate (E(2)B)-primed rats by 5ß-dihydroprogesterone (5ß-DHP), 5ß-pregnan-3ß-ol-20-one (5ß,3ß-Pgl), GnRH, PGE(2) and by manual flank/vaginocervical stimulation. These results suggest that the Src signaling system, by activating mitogen-activated protein kinases, participates in the facilitation of estrous behavior in E(2)B-primed rats induced by agents lacking affinity for the PR.


Asunto(s)
Dinoprostona/farmacología , Estradiol/farmacología , Estro/efectos de los fármacos , Hormona Liberadora de Gonadotropina/farmacología , Progestinas/farmacología , Conducta Sexual Animal/efectos de los fármacos , Familia-src Quinasas/fisiología , Animales , Cuello del Útero/efectos de los fármacos , Esquema de Medicación , Estradiol/administración & dosificación , Femenino , Estimulación Física , Progestinas/química , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Vagina/efectos de los fármacos , Vagina/fisiología , Familia-src Quinasas/metabolismo
8.
Brain Res ; 1474: 82-90, 2012 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-22824334

RESUMEN

Global ischemia arising during cardiac arrest or cardiac surgery causes highly selective, delayed death of hippocampal CA1 neurons. Phytoestrogens are naturally occurring plant-derived compounds that are present in the human diet and are considered selective estrogen receptor (ER) modulators. The phytoestrogen coumestrol is a potent isoflavonoid, with binding affinities for both ER-α and ER-ß that are comparable to those of 17 b-estradiol. The present study examined the hypothesis that coumestrol protects hippocampal neurons in ovariectomized rats in a model of cerebral global ischemia. Ovariectomized rats were subjected to global ischemia (10 min) or sham surgery and received a single intracerebroventricular or peripheral infusion of 20 µg of coumestrol, 20 µg of estradiol or vehicle 1h before ischemia or 0 h, 3h, 6h or 24h after reperfusion. Estradiol and coumestrol afforded significant neuroprotection in all times of administration, with the exception of estradiol given 24h after the ischemic insult. Animals received icv infusion of the broad-spectrum ER antagonist ICI 182,780 (50 µg) or vehicle into the lateral ventricle just before the E2 or coumestrol administration. The ER antagonist abolished estradiol protection, consistent with a role of classical ERs. In contrast, ICI 182,780 effected only partial reversal of the neuroprotective actions of coumestrol, suggesting that other cellular mediators in addition to classical ERs may be important. Additional research is needed to determine the molecular targets mediating the neuroprotective action of coumestrol and the therapeutic potential of this phytoestrogen in the mature nervous system.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Cumestrol/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Fitoestrógenos/administración & dosificación , Animales , Isquemia Encefálica/patología , Estradiol/administración & dosificación , Antagonistas de Estrógenos/farmacología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/patología , Inyecciones Intraventriculares , Neuronas/efectos de los fármacos , Neuronas/patología , Ovariectomía , Ratas , Ratas Wistar
9.
PLoS One ; 7(6): e38018, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22675505

RESUMEN

BACKGROUND: Transient global forebrain ischemia causes selective, delayed death of hippocampal CA1 pyramidal neurons, and the ovarian hormone 17ß-estradiol (E2) reduces neuronal loss in young and middle-aged females. The neuroprotective efficacy of E2 after a prolonged period of hormone deprivation is controversial, and few studies examine this issue in aged animals given E2 treatment after induction of ischemia. METHODOLOGY/PRINCIPAL FINDINGS: The present study investigated the neuroprotective effects of E2 administered immediately after global ischemia in aged female rats (15-18 months) after 6 months of hormone deprivation. We also used electrophysiological methods to assess whether CA1 synapses in the aging hippocampus remain responsive to E2 after prolonged hormone withdrawal. Animals were ovariohysterectomized and underwent 10 min global ischemia 6 months later. A single dose of E2 (2.25 µg) infused intraventricularly after reperfusion significantly increased cell survival, with 45% of CA1 neurons surviving vs 15% in controls. Ischemia also induced moderate loss of CA3/CA4 pyramidal cells. Bath application of 1 nM E2 onto brain slices derived from non-ischemic aged females after 6 months of hormone withdrawal significantly enhanced excitatory transmission at CA1 synapses evoked by Schaffer collateral stimulation, and normal long-term potentiation (LTP) was induced. The magnitude of LTP and of E2 enhancement of field excitatory postsynaptic potentials was indistinguishable from that recorded in slices from young rats. CONCLUSIONS/SIGNIFICANCE: The data demonstrate that 1) acute post-ischemic infusion of E2 into the brain ventricles is neuroprotective in aged rats after 6 months of hormone deprivation; and 2) E2 enhances synaptic transmission in CA1 pyramidal neurons of aged long-term hormone deprived females. These findings provide evidence that the aging hippocampus remains responsive to E2 administered either in vivo or in vitro even after prolonged periods of hormone withdrawal.


Asunto(s)
Envejecimiento/efectos de los fármacos , Isquemia Encefálica/fisiopatología , Estradiol/farmacología , Hipocampo/fisiopatología , Neuronas/patología , Ovariectomía , Transmisión Sináptica/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Estradiol/administración & dosificación , Femenino , Hipocampo/irrigación sanguínea , Hipocampo/efectos de los fármacos , Hipocampo/patología , Inyecciones Intraventriculares , Potenciación a Largo Plazo/efectos de los fármacos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos , Factores de Tiempo
10.
Behav Brain Res ; 232(1): 159-64, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22522024

RESUMEN

It is well established that estradiol (E2) decreases food intake and body weight in young female rats. However, it is not clear if female rats retain responsiveness to the anorexigenic effect of E2 during middle age. Because middle-aged females exhibit reduced responsiveness to E2, manifesting as a delayed and attenuated luteinizing hormone surge, it is plausible that middle-aged rats are less responsive to the anorexigenic effect of E2. To test this we monitored food intake in ovariohysterectomized young and middle-aged rats following E2 treatment. E2 decreased food intake and body weight to a similar degree in both young and middle-aged rats. Next, we investigated whether genes that mediate the estrogenic inhibition of food intake are similarly responsive to E2 by measuring gene expression of the anorexigenic genes corticotropin-releasing hormone (CRH), proopiomelanocortin (POMC), the long form of the leptin receptor (Lepr) and serotonin 2C receptors (5HT2CR) and the orexigenic genes agouti-related peptide (AgRP), neuropeptide Y (NPY), prepromelanin-concentrating hormone (pMCH) and orexin in the hypothalamus of young and middle-aged OVX rats treated with E2. As expected, E2 increased expression of all anorexigenic genes while decreasing expression of all orexigenic genes in young rats. Although CRH, 5HT2CR, Lepr, AgRP, NPY and orexin were also sensitive to E2 treatment in middle-aged rats, POMC and pMCH expression were not influenced by E2 in middle-aged rats. These data demonstrate that young and middle-aged rats are similarly sensitive to the anorexigenic effect of E2 and that most, but not all feeding-related genes retain sensitivity to E2.


Asunto(s)
Envejecimiento/psicología , Depresores del Apetito , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Estradiol/farmacología , Animales , Peso Corporal/efectos de los fármacos , ADN Complementario/biosíntesis , ADN Complementario/genética , Interpretación Estadística de Datos , Diestro/efectos de los fármacos , Estradiol/administración & dosificación , Receptor alfa de Estrógeno/biosíntesis , Femenino , Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ovariectomía , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Neuroendocrinology ; 96(2): 119-30, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22538356

RESUMEN

This review is intended to assess the state of current knowledge on the role of estrogen receptors (ERs) in the neuroprotective effects of estrogens in models for acute neuronal injury and death. We evaluate the overall evidence that estrogens are neuroprotective in acute injury and critically assess the role of ERα, ERß, GPR 30, and nonreceptor-mediated mechanisms in these robust neuroprotective effects of this ovarian steroid hormone. We conclude that all three receptors, as well as nonreceptor-mediated mechanisms can be involved in neuroprotection, depending on the model used, the level of estrogen administrated, and the mode of administration of the steroid. Also, the signaling pathways used by both ER-dependent and ER-independent mechanisms to exert neuroprotection are considered. Finally, further studies that are needed to parse out the relative contribution of receptor versus nonreceptor-mediated signaling are discussed.


Asunto(s)
Encefalopatías/tratamiento farmacológico , Estrógenos/metabolismo , Fármacos Neuroprotectores/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Encefalopatías/metabolismo , Estrógenos/farmacología , Humanos , Fármacos Neuroprotectores/farmacología
12.
Neuropeptides ; 46(1): 49-53, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22019256

RESUMEN

Intracerebroventricular (icv) administration of leptin facilitates lordosis behavior in ad libitum-fed, estrogen-primed rats. The cellular mechanism involved in this response is unknown. The present study tested the hypothesis that the nitric oxide-guanylyl cyclase, cGMP-dependent protein kinase (PKG) pathway is involved in the facilitation of lordosis behavior induced by the central administration of leptin. We tested the importance of the nitric oxide/cGMP pathway for lordosis stimulation by either icv infusion of a nitric oxide synthase inhibitor (L-NAME) or a nitric oxide-dependent, soluble guanylyl cyclase inhibitor (ODQ) 30 min before leptin administration (1 µg). This dose of leptin reliably induced lordosis behavior in ovariectomized estradiol benzoate treated rats. The lordosis induced by leptin at 1 and 2h after infusion was significantly reduced by the previous injection of either L-NAME or by ODQ. Intracerebroventricular infusion of the PKG inhibitor (KT5823) 30 min before leptin infusion, also significantly inhibited the lordosis behavior induced by leptin at 1 and 2h after hormone administration. These data support the hypothesis that the nitric oxide/cGMP/PKG pathway is involved in the facilitation of lordosis by leptin in estrogen-primed female rats.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Leptina/administración & dosificación , Óxido Nítrico/metabolismo , Conducta Sexual Animal , Animales , Carbazoles/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Femenino , Guanilato Ciclasa/farmacología , Leptina/farmacología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/fisiología , Ovariectomía , Postura , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/farmacología , Guanilil Ciclasa Soluble
13.
PLoS One ; 6(10): e26317, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22046273

RESUMEN

The brain contains numerous mononuclear phagocytes called microglia. These cells express the transmembrane tyrosine kinase receptor for the macrophage growth factor colony stimulating factor-1 (CSF-1R). Using a CSF-1R-GFP reporter mouse strain combined with lineage defining antibody staining we show in the postnatal mouse brain that CSF-1R is expressed only in microglia and not neurons, astrocytes or glial cells. To study CSF-1R function we used mice homozygous for a null mutation in the Csflr gene. In these mice microglia are >99% depleted at embryonic day 16 and day 1 post-partum brain. At three weeks of age this microglial depletion continues in most regions of the brain although some contain clusters of rounded microglia. Despite the loss of microglia, embryonic brain development appears normal but during the post-natal period the brain architecture becomes perturbed with enlarged ventricles and regionally compressed parenchyma, phenotypes most prominent in the olfactory bulb and cortex. In the cortex there is increased neuronal density, elevated numbers of astrocytes but reduced numbers of oligodendrocytes. Csf1r nulls rarely survive to adulthood and therefore to study the role of CSF-1R in olfaction we used the viable null mutants in the Csf1 (Csf1(op)) gene that encodes one of the two known CSF-1R ligands. Food-finding experiments indicate that olfactory capacity is significantly impaired in the absence of CSF-1. CSF-1R is therefore required for the development of microglia, for a fully functional olfactory system and the maintenance of normal brain structure.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Microglía/patología , Trastornos del Olfato/etiología , Receptor de Factor Estimulante de Colonias de Macrófagos/deficiencia , Animales , Encéfalo/patología , Ratones , Microglía/química , Vías Olfatorias , Receptor de Factor Estimulante de Colonias de Macrófagos/análisis , Receptor de Factor Estimulante de Colonias de Macrófagos/fisiología
14.
Endocrinology ; 152(11): 4276-87, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21914776

RESUMEN

Interactions between brain IGF-I receptors and estrogen receptors regulate female reproductive physiology and behavior. The present study investigated potential mechanisms by which IGF-I receptors in the neuroendocrine hypothalamus regulate GnRH neuronal activation and LH release in young and middle-aged female rats under estradiol (E2) positive feedback conditions. We infused vehicle, IGF-I, or JB-1, a selective antagonist of IGF-I receptors, into the third ventricle of ovariectomized female rats primed with E2 and progesterone or vehicle. In young females, blockade of IGF-I receptors attenuated the steroid hormone-induced LH surge, reduced the percent of GnRH neurons expressing c-fos on the day of the LH surge, and decreased the total number of neurons expressing c-fos in the preoptic area. Middle-aged females had fewer GnRH neurons expressing c-fos during the LH surge than young females, and the LH surge amplitude was attenuated. Infusion of an IGF-I dose previously shown to increase LH surge amplitude did not increase the percent of GnRH neurons expressing c-fos in middle-aged females. Brain IGF-I receptor blockade did not modify E2 induction of progestin receptor-immunoreactive neurons in the preoptic area, arcuate, or ventromedial hypothalamus of young rats. These findings indicate that brain IGF-I receptors are required for E2 activation of GnRH neurons in young rats and for robust GnRH release from axon terminals in middle-aged females. IGF-I likely exerts its effects by actions on E2-sensitive neurons that are upstream of GnRH neurons and terminals.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Hormona Luteinizante/sangre , Neuronas/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo , Factores de Edad , Animales , Recuento de Células , Estradiol/farmacología , Femenino , Hipotálamo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neuronas/metabolismo , Ovariectomía , Progesterona/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptores de Progesterona/metabolismo
15.
Neuropeptides ; 45(1): 63-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21112629

RESUMEN

Dose response curves for leptin facilitation of estrous behavior (lordosis and proceptivity) were made by infusing the peptide into the lateral ventricle (icv) of ovariectomized (ovx), ad libitum-fed rats injected 40h previously with 5µg of estradiol benzoate. Leptin doses of 1 and 3µg produced significant lordosis quotient at 60min post-injection, with maximal lordosis being displayed at 120min. Yet the intensity of lordosis was weak, and a high incidence of rejection behaviors was found. Moreover, leptin did not induce significant proceptive behaviors at any dose. The leptin doses of 1 and 3µg were selected for determining whether antide, a GnRH-1 receptor antagonist, or the progestin receptor antagonist RU486 could modify the lordosis response to leptin. Icv injection of either antide or RU486 1h before leptin significantly depressed leptin facilitation of lordosis. The results suggest that leptin stimulates lordosis by releasing GnRH, which in turn activates GnRH-1 and progestin receptors. The physiological role of leptin in the control of estrous behavior remains to be determined.


Asunto(s)
Estrógenos/farmacología , Leptina/farmacología , Lordosis , Receptores LHRH/antagonistas & inhibidores , Receptores de Progesterona/metabolismo , Conducta Sexual Animal/efectos de los fármacos , Animales , Femenino , Antagonistas de Hormonas/farmacología , Infusiones Intraventriculares , Masculino , Mifepristona/farmacología , Oligopéptidos/farmacología , Ovariectomía , Ratas , Ratas Sprague-Dawley
16.
Front Neuroendocrinol ; 32(3): 336-52, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21163293

RESUMEN

This review highlights our investigations into the neuroprotective efficacy of estradiol and other estrogenic agents in a clinically relevant animal model of transient global ischemia, which causes selective, delayed death of hippocampal CA1 neurons and associated cognitive deficits. We find that estradiol rescues a significant number of CA1 pyramidal neurons that would otherwise die in response to global ischemia, and this is true when hormone is provided as a long-term pretreatment at physiological doses or as an acute treatment at the time of reperfusion. In addition to enhancing neuronal survival, both forms of estradiol treatment induce measurable cognitive benefit in young animals. Moreover, estradiol and estrogen analogs that do not bind classical nuclear estrogen receptors retain their neuroprotective efficacy in middle-aged females deprived of ovarian hormones for a prolonged duration (8weeks). Thus, non-feminizing estrogens may represent a new therapeutic approach for treating the neuronal damage associated with global ischemia.


Asunto(s)
Congéneres del Estradiol/uso terapéutico , Estradiol/uso terapéutico , Isquemia/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Humanos , Isquemia/patología , Isquemia/fisiopatología
17.
Cancer Epidemiol Biomarkers Prev ; 19(10): 2421-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20841389

RESUMEN

Gliomas are the most common type of primary malignant brain tumor and have a very poor prognosis. Little is known, however, about the etiology of these tumors. Evidence from a number of sources suggests that endogenous steroid hormones may play a role in the development of gliomas. First, the descriptive epidemiology of glioma suggests a relative protection of females compared with males, particularly during the premenopausal years. Second, some gliomas and glioblastomas express estrogen receptors (ER), especially ERß, as well as aromatase, the enzyme responsible for the conversion of testosterone to estradiol, and possibly other steroid hormone receptors. Third, experimental studies indicate that glioblastomas transplanted into animals grow at a slower rate in females compared with males. Finally, experimental studies show that estradiol, 2-methoxyestradiol, and a number of selective estrogen receptor modulators inhibit proliferation of gliomas and induce cell death. These hormonal agonists and antagonists may act either through classical steroid hormone receptors or independently of such receptors. In view of these findings, further clinical, experimental, and epidemiologic studies are needed to elucidate the role of steroid hormone agonists and antagonists in the development and proliferation of glioma. If hormonal pathways are involved in gliomagenesis, this could eventually lead to the design of preventive strategies.


Asunto(s)
Glioma/etiología , Glioma/metabolismo , Hormonas/metabolismo , Neoplasias Hormono-Dependientes/etiología , Neoplasias Hormono-Dependientes/metabolismo , Esteroides/metabolismo , Animales , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Masculino
18.
Sci Eng Ethics ; 16(4): 753-62, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20585892

RESUMEN

The basic components of data management including data ownership, collection, selection, recording, analysis, storage, retention, destruction, and sharing. A number of important principles underlie best practices for each of these components; these include recording details such that another can repeat the experiment, keeping the data safe, managing storage in such a way as to facilitate easy retrieval for the period of time required by regulatory agencies and establishing data sharing principles with colleagues before collaborations begin. Experience as practicing scientists and teachers has aided in developing helpful strategies and approaches for communicating these principles, policies and practices to trainees and colleagues. We recommend didactic instruction focused by discipline, combined with the use of "teachable moments" in a student's career, as well as teaching principles versus rules, because changing methods of data collection and storage have implications for data management practices.


Asunto(s)
Comunicación , Control de Formularios y Registros/normas , Gestión de la Información/educación , Proyectos de Investigación , Enseñanza/métodos , Control de Formularios y Registros/métodos , Regulación Gubernamental , Gestión de la Información/métodos , Gestión de la Información/normas , Políticas , Investigación/educación , Estados Unidos
19.
Horm Behav ; 58(2): 223-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20307541

RESUMEN

This study tested the hypothesis that the Src/Raf/MAPK signaling pathway is involved in the facilitation of the lordosis and proceptive behaviors induced by progesterone (P) and its ring A-reduced metabolites in ovariectomized, estradiol-primed rats. Intraventricular (icv) infusion of PP2 (7.5, 15 and 30 microg), a Src kinase inhibitor, significantly depressed P-dependent estrous behavior (lordosis and proceptivity) in estradiol-primed rats. Icv infusion of 30 microg of PP2 also significantly attenuated estrous behavior induced by the ring A-reduced P metabolites 5 alpha-dihydroprogesterone (5 alpha-DHP) and 5 alpha-pregnan-3alpha-ol-20-one (allopregnanolone). PP2 did not inhibit estrous behavior induced by administration of high doses of estradiol alone to ovariectomized rats. We also assessed if the ventromedial hypothalamus (VMH) is one of the neural sites at which progestins activate Src signaling to facilitate estrous behavior. Bilateral administration of 15 microg of PP2 into the VMH inhibited the stimulation of both lordosis and proceptive behaviors elicited by subcutaneous P administration to estradiol-primed rats. These results suggest that progestins act through Src/Raf/MAPK signaling to initiate estrous behaviors in estrogen-primed rats. This event is one component of the cellular pathways leading to the display of estrous behaviors induced by P and its ring A-reduced metabolites in female rats.


Asunto(s)
Estradiol/metabolismo , Ciclo Estral/fisiología , Progestinas/metabolismo , Conducta Sexual Animal/fisiología , Familia-src Quinasas/metabolismo , 5-alfa-Dihidroprogesterona/metabolismo , Animales , Inhibidores Enzimáticos/farmacología , Ciclo Estral/efectos de los fármacos , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Modelos Neurológicos , Ovariectomía , Pregnanolona/metabolismo , Progesterona/metabolismo , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Conducta Sexual Animal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Familia-src Quinasas/antagonistas & inhibidores
20.
Brain Res ; 1321: 1-12, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20114038

RESUMEN

Global ischemia arising during cardiac arrest or cardiac surgery causes highly selective, delayed death of hippocampal CA1 neurons. Exogenous estradiol ameliorates global ischemia-induced neuronal death and cognitive impairment in male and female rodents. However, the molecular mechanisms by which a single acute injection of estradiol administered after the ischemic event intervenes in global ischemia-induced apoptotic cell death are unclear. Here we show that acute estradiol acts via the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling cascade to protect CA1 neurons in ovariectomized female rats. We demonstrate that global ischemia promotes early activation of glycogen synthase kinase-3beta (GSK3beta) and forkhead transcription factor of the O class (FOXO)3A, known Akt targets that are related to cell survival, and activation of caspase-3. Estradiol prevents ischemia-induced dephosphorylation and activation of GSK3beta and FOXO3A, and the caspase death cascade. These findings support a model whereby estradiol acts by activation of PI3K/Akt signaling to promote neuronal survival in the face of global ischemia.


Asunto(s)
Apoptosis/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Estradiol/farmacología , Estrógenos/farmacología , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Western Blotting , Isquemia Encefálica/patología , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Degeneración Nerviosa/prevención & control , Neuronas/metabolismo , Neuronas/patología , Ovariectomía , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...