Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros











Intervalo de año de publicación
1.
eNeuro ; 4(5)2017.
Artículo en Inglés | MEDLINE | ID: mdl-29098175

RESUMEN

New cells are added during both puberty and adulthood to hypothalamic regions that govern reproduction, homeostasis, and social behaviors, yet the functions of these late-born cells remain elusive. Here, we pharmacologically inhibited cell proliferation in ventricular zones during puberty or in adulthood and determined subsequent effects on the hormone-induced surge of luteinizing hormone (LH) in female rats. Initial neuroanatomical analyses focused on verifying incorporation, activation, and pharmacological inhibition of pubertally or adult born cells in the anteroventral periventricular nucleus (AVPV) of the hypothalamus because of the essential role of the AVPV in triggering the preovulatory LH surge in females. We first showed that approximately half of the pubertally born AVPV cells are activated by estradiol plus progesterone (P) treatment, as demonstrated by Fos expression, and that approximately 10% of pubertally born AVPV cells express estrogen receptor alpha (ERα). Next, we found that mitotic inhibition through intracerebroventricular (ICV) administration of cytosine ß-D-arabinofuranoside (AraC), whether during puberty or in adulthood, decreased the number of new cells added to the AVPV and the suprachiasmatic nucleus (SCN), and also blunted and delayed the hormone-induced LH surge. These studies do not prove, but are highly suggestive, that ongoing postnatal addition of new cells in periventricular brain regions, including the AVPV and SCN, may be important to the integrity of female reproduction.


Asunto(s)
Hipotálamo Anterior/citología , Hipotálamo Anterior/metabolismo , Hormona Luteinizante/metabolismo , Maduración Sexual/fisiología , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/metabolismo , Animales , Antimitóticos/farmacología , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Estradiol/administración & dosificación , Estradiol/metabolismo , Receptor alfa de Estrógeno/metabolismo , Femenino , Hipotálamo Anterior/efectos de los fármacos , Hipotálamo Anterior/crecimiento & desarrollo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Progesterona/administración & dosificación , Progesterona/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Receptores de Progesterona/metabolismo , Núcleo Supraquiasmático/efectos de los fármacos , Núcleo Supraquiasmático/crecimiento & desarrollo
2.
Am J Physiol Heart Circ Physiol ; 309(2): H335-44, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25980022

RESUMEN

The role of serotonin in the hemodynamic response to blood loss remains controversial. Caudal raphe serotonin neurons are activated during hypotensive hemorrhage, and their destruction attenuates sympathetic increases following blood loss in unanesthetized rats. Caudal raphe neurons provide serotonin-positive projections to the nucleus tractus solitarii (NTS), and disruption of serotonin-positive nerve terminals in the NTS attenuates sympathetic recovery following hemorrhage. Administration of 5-HT1A-receptor agonists following hemorrhage augments sympathetic-mediated increases in venous tone and tissue hypoxia. These findings led us to hypothesize that severe blood loss promotes activation of 5-HT1A receptors in the NTS, which facilitates sympathetic recovery and peripheral tissue perfusion. Here, we developed an adeno-associated viral vector encoding an efficacious small hairpin RNA sequence targeting the rat 5-HT1A receptor. Unanesthetized rats subjected to NTS injection of the anti-rat 5-HT1A small hairpin RNA-encoding vector 4 wk prior showed normal blood pressure recovery, but an attenuated recovery of renal sympathetic nerve activity (-6.4 ± 12.9 vs. 42.6 ± 15.6% baseline, P < 0.05) 50 min after 21% estimated blood volume withdrawal. The same rats developed increased tissue hypoxia after hemorrhage, as indicated by prolonged elevations in lactate (2.77 ± 0.5 vs. 1.34 ± 0.2 mmol/l, 60 min after start of hemorrhage, P < 0.05). 5-HT1A mRNA levels in the commissural NTS were directly correlated with renal sympathetic nerve activity (P < 0.01) and inversely correlated with lactate (P < 0.05) 60 min after start of hemorrhage. The data suggest that 5-HT1A receptors in the commissural NTS facilitate tissue perfusion after blood loss likely by increasing sympathetic-mediated venous return.


Asunto(s)
Barorreflejo , Hemorragia/metabolismo , Hipotensión/metabolismo , Riñón/inervación , Receptor de Serotonina 5-HT1A/metabolismo , Neuronas Serotoninérgicas/metabolismo , Núcleo Solitario/metabolismo , Sistema Nervioso Simpático/metabolismo , Animales , Barorreflejo/efectos de los fármacos , Presión Sanguínea , Volumen Sanguíneo , Modelos Animales de Enfermedad , Hemorragia/genética , Hemorragia/fisiopatología , Hipotensión/genética , Hipotensión/fisiopatología , Ácido Láctico/metabolismo , Masculino , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Receptor de Serotonina 5-HT1A/genética , Recuperación de la Función , Neuronas Serotoninérgicas/efectos de los fármacos , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Transducción de Señal , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/fisiopatología , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología , Factores de Tiempo
3.
Psychoneuroendocrinology ; 35(7): 1023-33, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20138435

RESUMEN

Estradiol regulates serotonin 1A (5-HT(1A)) receptor signaling. Since desensitization of 5-HT(1A) receptors may be an underlying mechanism by which selective serotonin reuptake inhibitors (SSRIs) mediate their therapeutic effects and combining estradiol with SSRIs enhances the efficacy of the SSRIs, it is important to determine which estrogen receptors are capable of desensitizating 5-HT(1A) receptor function. We previously demonstrated that selective activation of the estrogen receptor, GPR30, desensitizes 5-HT(1A) receptor signaling in rat hypothalamic paraventricular nucleus (PVN). However, since estrogen receptor-beta (ERbeta), is highly expressed in the PVN, we investigated the role of ERbeta in estradiol-induced desensitization of 5-HT(1A) receptor signaling. We first showed that a selective ERbeta agonist, diarylpropionitrile (DPN) has a 100-fold lower binding affinity than estradiol for GPR30. Administration of DPN did not desensitize 5-HT(1A) receptor signaling in rat PVN as demonstrated by agonist-stimulated hormone release. Second, we used a recombinant adenovirus containing ERbeta siRNAs to decrease ERbeta expression in the PVN. Reductions in ERbeta did not alter the estradiol-induced desensitization of 5-HT(1A) receptor signaling in oxytocin cells. In contrast, in animals with reduced ERbeta, estradiol administration, instead of producing desensitization, augmented the ACTH response to a 5-HT(1A) agonist. Combined with the results from the DPN treatment experiments, desensitization of 5-HT(1A) receptor signaling does not appear to be mediated by ERbeta in oxytocin cells, but that ERbeta, together with GPR30, may play a complex role in central regulation of 5-HT(1A)-mediated ACTH release. Determining the mechanisms by which estrogens induce desensitization may aid in the development of better treatments for mood disorders.


Asunto(s)
Estradiol/metabolismo , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/metabolismo , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Hormona Adrenocorticotrópica/sangre , Hormona Adrenocorticotrópica/metabolismo , Animales , Estradiol/farmacología , Femenino , Nitrilos/farmacología , Oxitocina/sangre , Oxitocina/metabolismo , Propionatos/farmacología , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/metabolismo
4.
Psychoneuroendocrinology ; 34 Suppl 1: S113-22, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19447561

RESUMEN

Decreasing levels of sex hormones with aging may have a negative impact on brain function, since this decrease is associated with the progression of neurodegenerative disorders, increased depressive symptoms and other psychological disturbances. Extensive evidence from animal studies indicates that sex steroids, in particular estradiol, are neuroprotective. However, the potential benefits of estradiol therapy for the brain are counterbalanced by negative, life-threatening risks in the periphery. A potential therapeutic alternative to promote neuroprotection is the use of selective estrogen receptor modulators (SERMs), which may be designed to act with tissue selectivity as estrogen receptor agonists in the brain and not in other organs. Currently available SERMs act not only with tissue selectivity, but also with cellular selectivity within the brain and differentially modulate the activation of microglia, astroglia and neurons. Finally, SERMs may promote the interaction of estrogen receptors with the neuroprotective signaling of growth factors, such as the phosphatidylinositol 3-kinase/glycogen synthase kinase 3 pathway.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Receptores de Estrógenos/agonistas , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Humanos , Modelos Biológicos , Neuroglía/metabolismo , Neuronas/metabolismo , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico
5.
Reproduction ; 135(4): 419-29, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18367504

RESUMEN

Neuron-to-glia, glia-to-neuron, and glia-to-glia communication are implicated in the modulation of neuronal activity and synaptic transmission relevant to reproduction. Glial cells play an important role in neuroendocrine regulation and participate in the sexual differentiation of neuronal connectivity of brain regions involved in the control of reproductive neuroendocrine output. During puberty, modifications in the morphology and chemistry of astrocytes and tanycytes in the hypothalamus and median eminence influence the maturation of the neuronal circuits controlling the secretion of GnRH. During adult reproductive life, the glial cells participate in the transient remodeling of neuronal connectivity in the preoptic area, the arcuate nucleus, the median eminence, and other brain regions involved in the control of reproduction. Gonadal hormones regulate glial plasticity by direct and indirect effects and regulate various other endocrine signals, local soluble factors and adhesion molecules that also affect glial function and glia-to-neuron communication. The glial cells, therefore, are central to the coordination of endocrine and local inputs that bring about neural plasticity and adapt reproductive capacity to homeostatic signals.


Asunto(s)
Hormona Liberadora de Gonadotropina/fisiología , Hipotálamo , Neuroglía/fisiología , Neuronas/fisiología , Sistemas Neurosecretores/fisiología , Reproducción/fisiología , Adulto , Comunicación Celular , Humanos , Plasticidad Neuronal
6.
Neurourol Urodyn ; 27(5): 440-5, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17957780

RESUMEN

AIM: Urinary incontinence affects a significant number of post-menopausal women. There is conflicting evidence whether voiding symptoms in these women are related to hypoestrogenism or aging itself. This neuroanatomical study was designed to determine whether a specific central nervous system (CNS) pathway that projects to the pontine micturition center (PMC, also known as "Barrington's nucleus") is estrogen sensitive in a rat model. METHODS: A fluorescent retrograde tracer was injected into the dorsal pontine tegmentum of adult female Sprague-Dawley rats to identify neurons in the medial preoptic area (MPA) that project to the PMC. Immunohistochemistry was performed using antibodies directed against estrogen receptor-alpha (ERalpha) and estrogen receptor-beta (ERbeta) to identify estrogen-sensitive neurons. The brain sections were examined using fluorescence microscopy to identify cells that project to the PMC (contain fluorescent tracer) and also express ER (are immunoreactive for ER). RESULTS: There are neurons in the MPA that are double labeled (contain fluorescent tracer and express ERalpha, but not ERbeta), showing that a subset of neurons projecting from the MPA to the PMC is estrogen sensitive. CONCLUSIONS: A subset of estrogen-sensitive neurons projects from the MPA to the PMC in rats, raising the possibility that indirect estrogenic regulation of forebrain neuronal function may modulate the micturition reflex. Future development of drugs that alter the function of this estrogen-sensitive CNS pathway may provide therapeutic strategies to treat post-menopausal incontinence.


Asunto(s)
Estrógenos/fisiología , Locus Coeruleus/fisiología , Vías Nerviosas/fisiología , Puente/fisiología , Área Preóptica/fisiología , Animales , Interpretación Estadística de Datos , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/metabolismo , Femenino , Colorantes Fluorescentes , Microscopía Fluorescente , Neuronas/metabolismo , Ovariectomía , Ratas , Ratas Sprague-Dawley , Estilbamidinas
7.
Expert Rev Endocrinol Metab ; 2(3): 387-397, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-30743812

RESUMEN

Although estradiol is a neuroprotective factor, estrogen therapy in older women increases the risk of adverse cognitive outcomes and poses additional peripheral risks, requiring careful use of estrogenic compounds as treatments for neurodegenerative conditions or neural injury. Potential alternatives to estrogen therapy to promote neuroprotection might include treatment with molecules that are able to interact with estrogen receptors, with alternative mechanisms of action, or with molecules that induce local estradiol synthesis in the brain, or a combination of all. However, before considering the broad clinical applications, more basic research is required to clarify the mechanisms of action and potential risks of some of these estrogen-based treatments.

8.
Endocrinology ; 146(10): 4340-8, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16002522

RESUMEN

Reproductive and behavioral functions of progesterone receptors (PRs) in males were assessed by examining consequences of PR gene deletion. Basal hormone levels were measured in male progesterone receptor knockout (PRKO) mice and compared to wild-type (WT) counterparts. RIA of serum LH, testosterone, and progesterone levels revealed no significant differences. Levels of FSH were moderately but significantly lower and inhibin levels were higher in PRKOs; these differences were not accompanied by gross differences in testicular weight or morphology. PRKOs exhibited significant alterations in sexual behavior. In initial tests PRKOs exhibited reduced latency to mount, compared with WT. In second sessions, PRKOs again showed a significantly reduced latency to mount and increased likelihood of achieving ejaculation. RU486 treatment in WT produced increased mount and intromission frequency and decreased latency to intromission. In anxiety-related behavior tests, PRKO mice exhibited intermediate anxiety levels, compared with WT, suggesting that enhanced sexual behavior in PRKOs is not secondary to reduced anxiety. Immunohistochemical analysis revealed significantly enhanced androgen receptor expression in the medial preoptic nucleus and bed nucleus of the stria terminalis of PRKO. We conclude that testicular development and function and homeostatic regulation of the hypothalamic-pituitary testicular axis are altered to a lesser extent by PR gene deletion. In contrast, PR appears to play a substantial role in inhibiting the anticipatory/motivational components of male sexual behavior in the mouse. The biological significance of this inhibitory mechanism and the extent to which it is mediated by reduced androgen receptor expression remain to be clarified.


Asunto(s)
Receptores Androgénicos/fisiología , Receptores de Progesterona/deficiencia , Receptores de Progesterona/genética , Conducta Sexual Animal , Animales , Ansiedad , Cartilla de ADN , Hormona Folículo Estimulante/sangre , Hormona Luteinizante/sangre , Masculino , Ratones , Ratones Noqueados , Mifepristona/farmacología , Actividad Motora , Tamaño de los Órganos , Reacción en Cadena de la Polimerasa , Progesterona/farmacología , Radioinmunoensayo , Recuento de Espermatozoides , Testículo/anatomía & histología
9.
J Neurosci ; 25(16): 4004-13, 2005 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-15843602

RESUMEN

In the hamster facial nerve injury paradigm, we have established that androgens enhance both functional recovery from facial nerve paralysis and the rate of regeneration in the adult, through intrinsic effects on the nerve cell body response to injury and via an androgen receptor (AR)-mediated mechanism. Whether these therapeutic effects of gonadal steroids encompass neuroprotection from axotomy-induced cell death is the focus of the present study. Virtually 100% of adult hamster facial motoneurons (FMNs) survive axotomy at the stylomastoid foramen (SMF), whereas, before postnatal day 15 (P15), developing FMNs undergo substantial axotomy-induced cell death. The first part of the present study focuses on determining when ARs are first expressed in developing hamster FMNs. Using AR immunocytochemistry, it was found that males express ARs by P2 and females by P4, which is the earliest demonstration of AR expression in mammalian motoneurons reported thus far in the literature. The second half examines the neuroprotective effects of testosterone propionate, 17-beta estradiol, and dihydrotestosterone on FMNs of P7 hamsters after facial nerve transection at the SMF. The results demonstrate that androgens and estrogens are equally able to rescue approximately 20% of FMNs from axotomy-induced cell death, with the effects permanent. This study is the first to investigate the effects of both androgens and estrogens on axotomy-induced cell death in one system and, with our previously published work, to validate the hamster FMN injury paradigm as a model of choice in the investigation of both neurotherapeutic and neuroprotective actions of gonadal steroids.


Asunto(s)
Traumatismos del Nervio Facial/tratamiento farmacológico , Traumatismos del Nervio Facial/patología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hormonas Esteroides Gonadales/farmacología , Neuronas Motoras/efectos de los fármacos , Factores de Edad , Animales , Animales Recién Nacidos , Axotomía/métodos , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/crecimiento & desarrollo , Tronco Encefálico/patología , Recuento de Células/métodos , Muerte Celular/efectos de los fármacos , Cricetinae , Dihidrotestosterona/farmacología , Dihidrotestosterona/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estradiol/farmacología , Estradiol/uso terapéutico , Femenino , Lateralidad Funcional , Regulación del Desarrollo de la Expresión Génica/fisiología , Hormonas Esteroides Gonadales/uso terapéutico , Inmunohistoquímica/métodos , Masculino , Mesocricetus , Neuronas Motoras/patología , Receptores Androgénicos/inmunología , Receptores Androgénicos/metabolismo , Factores Sexuales , Testosterona/farmacología , Testosterona/uso terapéutico
10.
Brain Res Brain Res Rev ; 48(2): 273-86, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15850667

RESUMEN

Proteins involved in the intramitochondrial trafficking of cholesterol, the first step in steroidogenesis, such as the steroidogenic acute regulatory protein (StAR) and the peripheral-type benzodiazepine receptor (PBR), are upregulated in the nervous system after injury. Accordingly, a local increase in the levels of steroids, such as pregnenolone and progesterone, is observed following traumatic injury in the brain and spinal cord. The expression and activity of aromatase, the enzyme that synthesizes estradiol, is also increased in injured brain areas and its inhibition results in an increased neurodegeneration. These findings suggest that an increase in steroidogenesis is part of an overall mechanism used by the nervous tissue to cope with neurodegenerative conditions. Neural steroidogenesis is the result of a coordinated interaction of neurons and glia. For example, after neural injury, there is an upregulation of StAR in neurons and of PBR in microglia and astroglia. Aromatase is expressed in neurons under basal conditions and is upregulated in reactive astrocytes after injury. Some of the steroids produced by glia are neuroprotective. Progesterone and progesterone derivatives produced by Schwann cells, promote myelin formation and the remyelination and regeneration of injured nerves. In the central nervous system, the steroids produced by glia regulate synaptic function, affect anxiety, cognition, sleep and behavior, and exert neuroprotective and reparative roles. In addition, glial cells are targets for steroids and mediate some of the effects of these molecules on neurons, including the regulation of survival and regeneration.


Asunto(s)
Lesiones Encefálicas/prevención & control , Hormonas Esteroides Gonadales/uso terapéutico , Neuroglía/fisiología , Neuronas/fisiología , Fármacos Neuroprotectores/uso terapéutico , Animales , Aromatasa/metabolismo , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/metabolismo , Comunicación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Modelos Biológicos , Neuroglía/clasificación
11.
Exp Gerontol ; 39(11-12): 1623-31, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15582278

RESUMEN

Sex steroids exert pleiotropic effects in the nervous system, preserving neural function and promoting neuronal survival. Therefore, the age-related decrease in sex steroids may have a negative impact on neural function. Progesterone, testosterone and estradiol prevent neuronal loss in the central nervous system in different experimental animal models of neurodegeneration. Furthermore, progesterone and its reduced derivatives dihydroprogesterone and tetrahydroprogesterone reduce aging-associated morphological abnormalities of myelin and aging-associated myelin fiber loss in rat peripheral nerves. However, the results from hormone replacement studies in humans are thus far inconclusive. A possible alternative to hormonal replacement therapy is to increase local steroidogenesis by neural tissues, which express enzymes for steroid synthesis and metabolism. Proteins involved in the intramitochondrial trafficking of cholesterol, the first step in steroidogenesis, such as the peripheral-type benzodiazepine receptor and the steroidogenic acute regulatory protein, are up-regulated in the nervous system after injury. Furthermore, steroidogenic acute regulatory protein expression is increased in the brain of 24-month-old rats compared with young adult rats. This suggests that brain steroidogenesis may be modified in adaptation to neurodegenerative conditions and to the brain aging process. Furthermore, recent studies have shown that local formation of estradiol in the brain, by the enzyme aromatase, is neuroprotective. Therefore, steroidogenic acute regulatory protein, peripheral-type benzodiazepine receptor and aromatase are attractive pharmacological targets to promote neuroprotection in the aged brain.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/fisiología , Hormonas Esteroides Gonadales/fisiología , Anciano , Animales , Aromatasa/metabolismo , Encéfalo/metabolismo , Femenino , Terapia de Reemplazo de Hormonas , Humanos , Masculino , Modelos Animales , Enfermedades Neurodegenerativas/metabolismo , Fosfoproteínas/metabolismo , Progesterona/metabolismo , Ratas , Receptores de GABA-A/metabolismo
12.
Neuroendocrinology ; 80(1): 31-41, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15385710

RESUMEN

The present study examined the effect of estradiol on hypothalamic serotonin-1A (5-HT(1A)) receptor signaling in female rats. We first examined the time-course effects of a single injection of the 5-HT(1A) receptor agonist (+/-)8-OH-DPAT (5, 15 or 30 min prior to decapitation), and dose response of (+)8-OH-DPAT (50, 100, 200 or 500 microg/kg, s.c.) on plasma hormones in ovariectomized rats that received a daily injection of beta-estradiol 3-benzoate (10 microg/day, s.c.) or vehicle (sesame oil) for 2 days. In vehicle- and estrogen-treated rats, the peak response of hormones occurred at 15 min after injection and the time-course of oxytocin and adrenocorticotropic hormone (ACTH) responses to an injection of 8-OH-DPAT were comparable. However, only the oxytocin response was reduced by estrogen treatment. A second experiment compared the ACTH and oxytocin responses with doses of 50 or 200 microg/kg, s.c. of (+)8-OH-DPAT vs. (+/-)8-OH-DPAT in ovariectomized rats that were treated with oil or beta-estradiol 3-benzoate (10 microg/day, s.c.) for 2 days. (+)8-OH-DPAT and (+/-)8-OH-DPAT produced a similar magnitude of increase in plasma levels of ACTH and oxytocin. Treatment with beta-estradiol 3-benzoate produced a significant and comparable reduction in the oxytocin response to the highest dose (200 microg/kg, s.c.) of both (+)8-OH-DPAT and (+/-)8-OH-DPAT but did not alter the ACTH response to either (+)8-OH-DPAT or (+/-)8-OH-DPAT. In the dose-response experiment, a dose of 50 microg/kg of (+)8-OH-DPAT produced a maximal increase in plasma levels of ACTH, while the maximal oxytocin response was achieved with a dose of 200 microg/kg, s.c. Treatment with beta-estradiol 3-benzoate reduced the maximal oxytocin response to (+)8-OH-DPAT (by 29%) but did not alter the ACTH response to any doses of (+)8-OH-DPAT. To examine potential mechanisms mediating the effects of estrogen on 5-HT(1A) receptor signaling, we measured the levels of Galpha(i), Galpha(o) and Galpha(z) proteins, which couple 5-HT(1A) receptors to their effector enzymes, in two subregions of the hypothalamus. The levels of Galpha(z) protein were reduced in the mediobasal hypothalamus (containing the ventromedial and arcuate nuclei), which mainly expresses estrogen receptor-alpha, but not in the paraventricular hypothalamus, which mainly expresses estrogen receptor-beta. Estradiol reduced the levels of Galpha(i2) and Galpha(i3 )proteins in both hypothalamic regions but did not affect Galpha(i1) levels in either area. Combined, the data suggest that racemic and stereoselective 8-OH-DPAT have similar neuroendocrine effects and that both estrogen receptor-alpha and estrogen receptor-beta mediate the reduction in levels of Galpha(i2,3) proteins.


Asunto(s)
Estradiol/farmacología , Proteínas de Unión al GTP/efectos de los fármacos , Hipotálamo/metabolismo , Oxitocina/efectos de los fármacos , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Receptor de Serotonina 5-HT1A/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralin/química , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Hormona Adrenocorticotrópica/sangre , Hormona Adrenocorticotrópica/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Femenino , Proteínas de Unión al GTP/metabolismo , Hipotálamo/efectos de los fármacos , Immunoblotting , Ovariectomía , Oxitocina/sangre , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/metabolismo , Antagonistas de la Serotonina/química , Antagonistas de la Serotonina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Estereoisomerismo , Factores de Tiempo
13.
J Comp Neurol ; 450(3): 256-71, 2002 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-12209854

RESUMEN

Estrogens and androgens can protect neurons from death caused by injury to the central nervous system. Astrocytes and microglia are major players in events triggered by neural lesions. To determine whether glia are direct targets of estrogens or androgens after neural insults, steroid receptor expression in glial cells was assessed in two different lesion models. An excitotoxic injury to the hippocampus or a stab wound to the parietal cortex and hippocampus was performed in male rats, and the resultant expression of steroid receptors in glial cells was assessed using double-label immunohistochemistry. Both lesions induced the expression of estrogen receptors (ERs) and androgen receptors (ARs) in glial cells. ERalpha was expressed in astrocytes immunoreactive (ERalpha-ir) for glial fibrillary acidic protein or vimentin. AR immunoreactivity colocalized with microglial markers, such as Griffonia simplicifolia lectin-1 or OX-6. The time course of ER and AR expression in glia was studied in the stab wound model. ERalpha-ir astrocytes and AR-ir microglia were observed 3 days after lesion. The number of ERalpha-ir and AR-ir glial cells reached a maximum 7 days after lesion and returned to low levels by 28 days postinjury. The studies of ERbeta expression in glia were inconclusive; different results were obtained with different antibodies. In sum, these results suggest that reactive astrocytes and reactive microglia are a direct target for estrogens and androgens, respectively.


Asunto(s)
Lesiones Encefálicas/metabolismo , Degeneración Nerviosa/metabolismo , Regeneración Nerviosa/fisiología , Neuroglía/metabolismo , Lectinas de Plantas , Ratas Wistar/crecimiento & desarrollo , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/metabolismo , Gliosis/patología , Gliosis/fisiopatología , Inmunohistoquímica , Ácido Kaínico , Lectinas/metabolismo , Masculino , Microglía/citología , Microglía/metabolismo , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Ratas , Ratas Wistar/lesiones , Ratas Wistar/metabolismo , Vimentina/metabolismo
14.
Neurotox Res ; 4(3): 235-45, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12829404

RESUMEN

Accumulated clinical and basic evidence suggests that gonadal steroids affect the onset and progression of several neurodegenerative diseases and schizophrenia, and the recovery from traumatic neurological injury such as stroke. Thus, our view on gonadal hormones in neural function must be broadened to include not only their function in neuroendocrine regulation and reproductive behaviors, but also to include a direct participation in response to degenerative disease or injury. Recent findings indicate that the brain up-regulates both estrogen synthesis and estrogen receptor expression at sites of injury. Genetic or pharmacological inactivation of aromatase, the enzyme involved in estrogen synthesis, indicates that the induction of this enzyme in the brain after injury has a neuroprotective role. Some of the mechanisms underlying the neuroprotective effects of estrogen may be independent of the classically defined nuclear estrogen receptors (ERs). Other neuroprotective effects of estrogen do depend on the classical nuclear ERs, through which estrogen alters expression of estrogen responsive genes that play a role in apoptosis, axonal regeneration, or general trophic support. Yet another possibility is that non-classical ERs in the membrane or cytoplasm alter phosphorylation cascades, such as those involved in the signaling of insulin-like growth factor-1 (IGF-1). Indeed, ERs and IGF-1 receptor interact in the activation of PI3K and MAPK signaling cascades and in the promotion of neuroprotection. The decrease in estrogen and IGF-1 levels with aging may thus result in an increased risk for neuronal pathological alterations after different forms of brain injury.

15.
Neuroendocrinology ; 76(6): 348-56, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12566942

RESUMEN

The present study investigated the effects of long-term estradiol withdrawal (ovariectomy) on hypothalamic serotonin-1A (5-HT(1A)) receptor signaling. Changes in neuroendocrine responses to the 5-HT(1A) agonist 8-OH-DPAT and levels of G(z) protein in the hypothalamus were used to examine 5-HT(1A) receptor signaling. Five days following ovariectomy, rats received daily injections of either 2 microg of beta-estradiol 3-benzoate or vehicle (subcutaneously) for 2, 4 or 14 days. Twenty-four hours after the last injection, and 15 min prior to sacrifice, rats were injected with (+/-)8-OH-DPAT (50 micro;g/kg, s.c.) or saline. Estradiol treatment did not alter basal corticotropin (ACTH) or oxytocin levels. Injection of (+/-)8-OH-DPAT produced significant increases in plasma ACTH and oxytocin levels. In the vehicle-treated rats, hormone responses to 8-OH-DPAT were enhanced in rats that received injections for 14 days compared with rats that received injections for either 2 or 4 days. Estradiol treatment for 4 or 14 days blunted this enhanced ACTH response to 8-OH-DPAT, whereas the oxytocin response to 8-OH-DPAT was only blunted after 14 daily injections of beta-estradiol 3-benzoate. The treatment with beta-estradiol 3-benzoate (2 microg/rat) did not reduce membrane-associated G(z) protein levels in the paraventricular nucleus of the hypothalamus. Hence, the inhibitory influence of a low dose of beta-estradiol 3-benzoate on 5-HT(1A) receptor signaling in the hypothalamus is not accompanied by a change in the levels of G(z) protein in the paraventricular hypothalamic nucleus. Results from the present study indicate a supersensitivity of 5-HT(1A) receptors after withdrawal of estradiol and suggest that estradiol suppresses 5-HT(1A) receptor signaling.


Asunto(s)
Estradiol/análogos & derivados , Estradiol/metabolismo , Hipotálamo/metabolismo , Receptores de Serotonina/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Hormona Adrenocorticotrópica/sangre , Hormona Adrenocorticotrópica/efectos de los fármacos , Animales , Esquema de Medicación , Estradiol/administración & dosificación , Estradiol/sangre , Estradiol/farmacología , Femenino , Proteínas de Unión al GTP/metabolismo , Hipotálamo/efectos de los fármacos , Immunoblotting , Ovariectomía , Oxitocina/sangre , Oxitocina/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Receptores de Serotonina 5-HT1 , Agonistas de Receptores de Serotonina/farmacología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
16.
J Steroid Biochem Mol Biol ; 83(1-5): 211-7, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12650718

RESUMEN

In the brain, as in other tissues, estradiol interacts with growth factors. One of the growth factors that is involved in the neural actions of estradiol is insulin-like growth factor-I (IGF-I). Estradiol and IGF-I cooperate in the central nervous system to regulate neuronal development, neural plasticity, neuroendocrine events and the response of neural tissue to injury. The precise molecular mechanisms involved in these interactions are still not well understood. In the central nervous system there is abundant co-expression of estrogen receptors (ERs) and IGF-I receptors (IGF-IRs) in the same cells. Furthermore, the expression of estrogen receptors and IGF-I receptors in the brain is cross-regulated. In addition, using specific antibodies for the phosphorylated forms of extracellular-signal regulated kinase (ERK) 1 and ERK2 and Akt/protein kinase B (Akt/PKB) it has been shown that estradiol affects IGF-I signaling pathways in the brain. Estradiol treatment results in a dose-dependent increase in the phosphorylation of ERK and Akt/PKB in the brain of adult ovariectomized rats. In addition, estradiol and IGF-I have a synergistic effects on the activation of Akt/PKB in the adult rat brain. These findings suggest that estrogen effects in the brain may be mediated in part by the activation of the signaling pathways of the IGF-I receptor.


Asunto(s)
Encéfalo/metabolismo , Estrógenos/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Animales , Diferenciación Celular , Sistema Nervioso Central/metabolismo , Activación Enzimática , Gonadotropinas/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , Unión Proteica , Receptores de Estrógenos/metabolismo , Transducción de Señal , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA