Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Metab ; 3(4): 338, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24944888
2.
Mol Psychiatry ; 19(7): 752-61, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24732669

RESUMEN

The brain receives and integrates environmental and metabolic information, transforms these signals into adequate neuronal circuit activities, and generates physiological behaviors to promote energy homeostasis. The responsible neuronal circuitries show lifetime plasticity and guaranty metabolic health and survival. However, this highly evolved organization has become challenged nowadays by chronic overload with nutrients and reduced physical activity, which results in an ever-increasing number of obese individuals worldwide. Research within the last two decades has aimed to decipher the responsible molecular and cellular mechanisms for regulation of the hypothalamic melanocortin neurons, which have a key role in the control of food intake and energy metabolism. This review maps the central connections of the melanocortin system and highlights its global position and divergent character in physiological and pathological metabolic events. Moreover, recently uncovered molecular and cellular processes in hypothalamic neurons and glial cells that drive plastic morphological and physiological changes in these cells, and account for regulation of food intake and energy metabolism, are brought into focus. Finally, potential functional interactions between metabolic disorders and psychiatric diseases are discussed.


Asunto(s)
Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Hipotálamo/fisiología , Melanocortinas/fisiología , Proopiomelanocortina/fisiología , Proteína Relacionada con Agouti/fisiología , Animales , Humanos , Hipotálamo/fisiopatología , Trastornos Mentales/fisiopatología , Modelos Neurológicos , Neuroglía/fisiología , Neuronas/fisiología , Neuropéptido Y/fisiología , Orgánulos/fisiología
3.
Neuroscience ; 192: 500-6, 2011 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-21699961

RESUMEN

Ghrelin, an orexigenic hormone produced by the stomach, increases food intake and enhances the locomotor and rewarding effects of cocaine. Consistent with these behavioral effects, ghrelin increases dopamine cell activity in the mesolimbic system resulting in elevated levels of dopamine release and turnover in target regions such as the ventral striatum. In the current study, we examined the psychostimulant effects of acute and daily cocaine in mice with targeted deletion of the ghrelin gene (ghrelin knockout; KO) and that of their wild-type (WT) littermates. We hypothesized that ghrelin-KO mice would be hyporesponsive to the effects of cocaine as reflected in attenuated locomotor activity following both acute and chronic injections, and that this would be correlated with striatal dopamine and dopamine metabolite concentrations. Results show that the locomotor stimulating effect of cocaine (10 mg/kg) was decreased in ghrelin-KO mice as compared with their WT littermates. In addition, repeated daily injection of cocaine resulted in gradual increases in locomotor activity in WT mice, an effect that was attenuated in ghrelin-KO mice. These behavioral effects were correlated with changes in dopamine utilization in the striatum of WT mice that were not seen in ghrelin-KO mice unless these were pretreated with ghrelin. These data suggest that ghrelin is important for normal function of the mesolimbic dopaminergic system, potentially modulating both dopamine release and reuptake.


Asunto(s)
Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Ghrelina/metabolismo , Actividad Motora/efectos de los fármacos , Animales , Dopamina/metabolismo , Ghrelina/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados
4.
Am J Physiol Regul Integr Comp Physiol ; 300(6): R1352-62, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21411766

RESUMEN

Maintenance of reduced body weight in lean and obese human subjects results in the persistent decrease in energy expenditure below what can be accounted for by changes in body mass and composition. Genetic and developmental factors may determine a central nervous system (CNS)-mediated minimum threshold of somatic energy stores below which behavioral and metabolic compensations for weight loss are invoked. A critical question is whether this threshold can be altered by environmental influences and by what mechanisms such alterations might be achieved. We examined the bioenergetic, behavioral, and CNS structural responses to weight reduction of diet-induced obese (DIO) and never-obese (CON) C57BL/6J male mice. We found that weight-reduced (WR) DIO-WR and CON-WR animals showed reductions in energy expenditure, adjusted for body mass and composition, comparable (-10-15%) to those seen in human subjects. The proportion of excitatory synapses on arcuate nucleus proopiomelanocortin neurons was decreased by ∼50% in both DIO-WR and CON-WR mice. These data suggest that prolonged maintenance of an elevated body weight (fat) alters energy homeostatic systems to defend a higher level of body fat. The synaptic changes could provide a neural substrate for the disproportionate decline in energy expenditure in weight-reduced individuals. This response to chronic weight elevation may also occur in humans. The mouse model described here could help to identify the molecular/cellular mechanisms underlying both the defense mechanisms against sustained weight loss and the upward resetting of those mechanisms following sustained weight gain.


Asunto(s)
Peso Corporal/fisiología , Encéfalo/anatomía & histología , Metabolismo Energético/fisiología , Homeostasis/fisiología , Aumento de Peso/fisiología , Pérdida de Peso/fisiología , Animales , Núcleo Arqueado del Hipotálamo/anatomía & histología , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/fisiología , Composición Corporal/fisiología , Peso Corporal/efectos de los fármacos , Encéfalo/fisiología , Restricción Calórica , Grasas de la Dieta/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Neuronas/citología , Neuronas/fisiología , Sinapsis/fisiología
5.
Neuroscience ; 171(4): 1032-40, 2010 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-20923696

RESUMEN

Obesity is associated with increased risk of diabetes, cardiovascular disease and several types of cancers. The hypothalamus is a region of the brain critical in the regulation of body weight. One of the critical and best studied hypothalamic circuits is comprised of the melanocortinergic orexigenic agouti-related protein (AgRP) and anorexigenic α-melanocyte stimulating hormone (α-MSH) neurons. These neurons project axons to the same hypothalamic target neurons and balance each other's activity leading to body weight regulation. We previously showed that the brain proteoglycan syndecan-3 regulates feeding behavior and body weight, and syndecan-3 null (SDC-3(-/-)) mice are lean and obesity resistant. Here we show that the melanocortin agonist Melanotan II (MTII) potently suppresses food intake and activates the hypothalamic paraventricular nuclei (PVN) in SDC-3(-/-) mice based on c-fos immunoreactivity. Interestingly, we determined that the AgRP neuropeptide is reduced in the PVN of SDC-3(-/-) mice compared to wild type mice. In contrast, neuropeptide Y, coexpressed in the AgRP neuron, is not differentially expressed nor is the counteracting neuropeptide α-MSH. These findings are unprecedented and indicate that AgRP protein localization can be selectively regulated within the hypothalamus resulting in altered neuropeptide response and tone.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Ingestión de Alimentos/genética , Regulación de la Expresión Génica/genética , Transducción de Señal/fisiología , Sindecano-3/deficiencia , Análisis de Varianza , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Privación de Alimentos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuropéptido Y/farmacología , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
6.
Neuroscience ; 164(2): 351-9, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19666088

RESUMEN

Ghrelin, an orexigenic hormone produced by the stomach, is secreted in anticipation of scheduled meals and in correlation with anticipatory locomotor activity. We hypothesized that ghrelin is directly implicated in stimulating locomotor activity in anticipation of scheduled meals. To test this hypothesis, we observed 24 h patterns of locomotor activity in mice with targeted mutations of the ghrelin receptor gene (GHSR KO) and wild-type littermates, all given access to food for 4 h daily for 14 days. While wild type (WT) and GHSR KO mice produced increases in anticipatory locomotor activity, anticipatory locomotor activity in GHSR KO mice was attenuated (P<0.05). These behavioral measures correlated with attenuated levels of Fos immunoreactivity in a number of hypothalamic nuclei from GHSR KO placed on the same restricted feeding schedule for 7 days and sacrificed at ZT4. Interestingly, seven daily i.p. ghrelin injections mimicked hypothalamic Fos expression patterns to those seen in mice under restricted feeding schedules. These data suggest that ghrelin acts in the hypothalamus to augment locomotor activity in anticipation of scheduled meals.


Asunto(s)
Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Actividad Motora/fisiología , Receptores de Ghrelina/metabolismo , Animales , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Ghrelina/metabolismo , Locomoción/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Fotoperiodo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Radioinmunoensayo , Receptores de Ghrelina/deficiencia , Receptores de Ghrelina/genética , Factores de Tiempo
7.
Int J Obes (Lond) ; 30 Suppl 1: S30-2, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16570102

RESUMEN

Presentations in this symposium addressed effects and modes of action of endocannabinoids in various tissues in relation to metabolic disorders. Endocannabinoids are produced and exert their effect in various brain sites, including the mesolimbic reward circuitry and the hypothalamus. Both of these regions have direct ties to energy metabolism regulation, particularly food intake and energy expenditure. These data clearly suggest that the observed beneficial effects of CB1 (cannabinoid receptor 1) receptor antagonists on obesity may be related to the central endocannabinoid system. On the other hand, data presented on cannabinoid action in the liver and white adipose tissues clearly indicate that CB1-mediated events in affecting metabolic phenotype may occur in peripheral tissues as well. This together with the reported results from human trials on CB1 antagonists showing that the initial anorectic effect of rimonabant is diminished after the first weeks while longer lasting weight loss is achieved do indicate that peripheral action of cannabinoids are very important in body weight regulation. Should this hold true in the long run, antagonizing CB1 receptors with compound not crossing the blood-brain barrier could revolutionize pharmaceutical approaches to obesity by offering a tool that short cuts the central nervous system.


Asunto(s)
Tejido Adiposo/metabolismo , Encéfalo/metabolismo , Moduladores de Receptores de Cannabinoides/fisiología , Metabolismo Energético , Animales , Depresores del Apetito/uso terapéutico , Regulación del Apetito , Tolerancia a Medicamentos , Homeostasis , Humanos , Hígado/metabolismo , Obesidad/tratamiento farmacológico , Piperidinas/uso terapéutico , Pirazoles/uso terapéutico , Receptor Cannabinoide CB1/antagonistas & inhibidores , Rimonabant
8.
Neuroscience ; 137(4): 1447-61, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16387447

RESUMEN

Uncoupling proteins in the inner mitochondrial membrane uncouples oxidative phosphorylation from ATP synthesis. It has been suggested that these proteins are involved in thermogenesis as well as in the regulation of reactive oxygen species production in the mitochondria. The present work was conducted to investigate the localization of the uncoupling protein 2-like immunoreactivity (uncoupling protein 2/3 immunoreactivity) in the main catecholaminergic projection fields in the rat brain as well as in the areas of the dopaminergic and noradrenergic nerve cell groups. In particular, the relationships of tyrosine hydroxylase, dopamine beta-hydroxylase and uncoupling protein 2/3 immunoreactivity were assessed by double immunolabeling and confocal laser microscopy analysis associated with computer-assisted image analysis. Uncoupling protein 2/3 immunoreactivity was observed in discrete dopaminergic terminals in the nucleus accumbens and in the cerebral cortex whereas it was found in scattered noradrenergic terminals in the caudate putamen and Islands of Calleja Magna. One interesting finding was that uncoupling protein 2/3 immunoreactivity together with tyrosine hydroxylase immunoreactivity in the shell of nucleus accumbens was observed surrounding the previously characterized D1 receptor rich nerve cell column system characterized by a relative lack of tyrosine hydroxylase immunoreactivity. Moreover, in animal models of dopaminergic pathway degeneration, plastic changes in uncoupling protein 2/3 terminals have been shown in the cerebral cortex and striatum as seen from the increased size and intensity of uncoupling protein 2/3 immunoreactivity of their varicosities. Taken together, these findings open up the possibility that uncoupling protein 2/3 could play an important role modulating the dopaminergic and noradrenergic neurotransmission within discrete brain regions.


Asunto(s)
Encéfalo/fisiología , Proteínas Portadoras/fisiología , Dopamina/fisiología , Proteínas de Transporte de Membrana/fisiología , Proteínas Mitocondriales/fisiología , Neuronas/fisiología , Norepinefrina/fisiología , Vías Aferentes/fisiología , Animales , Femenino , Inmunohistoquímica , Canales Iónicos , Microscopía Confocal , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/fisiología , Proteína Desacopladora 2 , Proteína Desacopladora 3
9.
J Neural Transm (Vienna) ; 112(1): 65-76, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15599605

RESUMEN

This review focuses on transmitter-receptor mismatches in the brain, which is one of the hallmarks of the Volume Transmission (VT) concept, and how this phenomenon may be related to local temperature gradients created by brain uncoupling protein 2 (UCP2), which uncouples oxidative phosphorylation from ATP synthesis, hereby generating heat. Recent studies on transmitter-receptor mismatches have revealed dopamine and opioid peptide receptor mismatches in the intercalated islands of the amygdala, which are GABAergic cell clusters regulating amygdaloid output. Such mismatches have also been found in regions belonging to the extended amygdala and the nucleus accumbens shell. Now substantial UCP2 immunoreactivity has been found within the above transmitter-receptor mismatch regions, suggesting that UCP2 may enhance diffusion and convection of DA and opioid peptides in such regions by generation of local temperature gradients, thereby contributing to a dynamic regulation of VT.


Asunto(s)
Química Encefálica/fisiología , Encéfalo/fisiología , Catecolaminas/fisiología , Proteínas de Transporte de Membrana/fisiología , Proteínas Mitocondriales/fisiología , Péptidos Opioides/fisiología , Transmisión Sináptica/fisiología , Animales , Encéfalo/citología , Humanos , Canales Iónicos , Proteína Desacopladora 2
10.
Mol Psychiatry ; 9(5): 531-8, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14569273

RESUMEN

Many anxiety disorders, as well as major depressive disorder (MDD), are at least twice as prevalent in women as in men, but the neurobiological basis of this discrepancy has not been well studied. MDD is often precipitated by exposure to uncontrollable stress, and is frequently characterized by abnormal or disrupted prefrontal cortex (PFC) function. In animals, exposure to stress has been shown to cause PFC dysfunction, but sex differences in this effect have not been investigated. The present study tested male and female rats on a PFC-dependent working memory task after administration of FG7142, a benzodiazepine inverse agonist that activates stress systems in the brain. Female rats were impaired by lower doses than males during proestrus (high estrogen), but not during estrus (low estrogen). Similarly, ovariectomized females showed increased stress sensitivity only after estrogen replacement. These results suggest that estrogen amplifies the stress response in PFC, which may increase susceptibility to stress-related disorders.


Asunto(s)
Estrógenos/fisiología , Corteza Prefrontal/fisiopatología , Caracteres Sexuales , Estrés Psicológico/fisiopatología , Animales , Carbolinas/toxicidad , Trastorno Depresivo Mayor/fisiopatología , Trastorno Depresivo Mayor/psicología , Susceptibilidad a Enfermedades , Terapia de Reemplazo de Estrógeno , Estro , Femenino , Habituación Psicofisiológica , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/etiología , Trastornos de la Memoria/fisiopatología , Ovariectomía , Proestro , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/inducido químicamente
11.
Neuroscience ; 117(4): 791-4, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12654332

RESUMEN

It has been shown that gonadal steroids have the capacity to induce synaptic plasticity in certain areas of the nervous system. Previously we have demonstrated that due to the effect of estradiol there is a transient decrease in the number of GABAergic axo-somatic synapses in the arcuate nucleus. By using systemic application of the tracer Fluorogold we retrogradely labeled a subpopulation of arcuate neurons that project to the median eminence. We than applied the disector method for synapse quantification and found that these "hypophysiotropic neurons" receive less axo-somatic inputs. We found that 17beta-estradiol induced a decrease in the numerical density of axo-somatic contacts of these retrogradely-labeled neoroendocrine cells. Our data support the hypothesis that the hormonally driven morphological synaptic plasticity is neuron specific within the arcuate nucleus and plays a decisive role in the regulation of anterior pituitary.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Estradiol/metabolismo , Vías Nerviosas/metabolismo , Plasticidad Neuronal/fisiología , Terminales Presinápticos/metabolismo , Estilbamidinas , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/ultraestructura , Recuento de Células , Estradiol/farmacología , Femenino , Colorantes Fluorescentes , Inmunohistoquímica , Eminencia Media/fisiología , Microscopía Electrónica , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/ultraestructura , Plasticidad Neuronal/efectos de los fármacos , Adenohipófisis/fisiología , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/ultraestructura , Ratas , Ratas Sprague-Dawley
12.
Int J Obes Relat Metab Disord ; 27(4): 433-42, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12664076

RESUMEN

OBJECTIVE: To test the hypothesis that either uncoupling protein-2 UCP2 or UCP3 or both together influence obesity and inflammation in transgenic mice. DESIGN: We generated 12 lines of transgenic mice for both human UCP2 and 3 using native promoters from a human bacterial artificial chromosome (BAC) clone. The BAC expresses no genes other than UCP2 and 3. Mice used for experiments are N4 or higher of backcross to C57BL/6J (B6). Each experiment used transgenic mice and their nontransgenic littermates. RESULTS: Northern blots confirmed expression on human UCP2 in adipose and spleen, while human UCP3 expression was detectable in gastrocnemius muscle. Western blots demonstrated a four-fold increase of UCP2 protein in spleens of Line 32 transgenic animals. Heterozygous mice of four lines showing expression of human UCP2 in spleen were examined for obesity phenotypes. There were no significant differences between Lines 1 and 32, but female transgenics of both lines had significantly smaller femoral fat depots than the control (littermate) mice (P=0.015 and 0.005, respectively). In addition, total fat of transgenic females was significantly less in Line 1 (P=0.05) and almost significantly different in Line 32 (P=0.06). Male Line 1 mice were leaner (P=0.04) while male Line 32 mice were almost significantly leaner (P=0.06). Heterozygous mice of Lines 35 and 44 showed no significant differences from the nontransgenic littermate controls. Effects of the UCP2/UCP3 transgene on obesity in Line 32 mice were confirmed by crossing transgenic mice with the B6.Cg-Ay agouti obese mice. B6.Cg-Ay carrying the UCP2/UCP3 transgene from Line 32 were significantly leaner than nontransgenic B6.Cg-Ay mice. Line 32 UCP2/UCP3 transgenics showed increased hypothalamic Neuropeptide (NPY) levels and food intake, with reduced spontaneous physical activity. Transgenic baseline interleukin4 (IL-4) and interleukin6 (IL-6) levels were low with lower or later increases after endotoxin injection compared to wild-type littermates. Endotoxin-induced fever was also diminished in transgenic male animals. Low-density lipoprotein (LDL) cholesterol levels were significantly higher in both Line 1 and 32 transgenics (P=0.05 and 0.001, respectively) after they had been placed on a moderate fat-defined diet containing 32% of calories from fat for 5 weeks. CONCLUSION: Moderate overexpression of UCP2 and 3 reduced fat mass and increased LDL cholesterol in two independent lines of transgenic mice. Thus, the reduced fat mass cannot be due to insertional mutagenesis since virtually identical fat pad weights and masses were observed with the two independent lines. Line 32 mice also have altered inflammation and mitochondrial function. We conclude that UCP2 and/or 3 have small but significant effects on obesity in mice, and that their mechanism of action may include alterations of metabolic rate.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Transporte de Membrana , Proteínas Mitocondriales , Obesidad/metabolismo , Proteínas/metabolismo , Tejido Adiposo/metabolismo , Animales , Metabolismo Basal , Northern Blotting , Western Blotting , Temperatura Corporal/fisiología , Proteínas Portadoras/genética , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Ingestión de Energía , Regulación de la Expresión Génica/genética , Frecuencia Cardíaca/fisiología , Inflamación/fisiopatología , Canales Iónicos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Obesidad/genética , Proteínas/genética , Proteína Desacopladora 2 , Proteína Desacopladora 3
13.
Exp Gerontol ; 38(1-2): 53-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12543261

RESUMEN

Unlike primates who undergo ovarian failure and loss of sex steroids at the end of reproduction, aging rodents undergo constant vaginal estrus followed by constant diestrus and finally anestrus, which indicates the absence of responsive ovarian follicles. The latter state is analogous to menopause in women. The timing of the appearance of constant estrus is determined by many factors including estrogen exposure in the brain during development and the number of times that the animal gets pregnant. The chief site of this reproductive aging in rat brains is the arcuate nucleus of the hypothalamus. The transition from normal cycles to constant estrus parallels the females' gradually decreased ability to respond to administered estradiol with a cycle of inhibition followed by disinhibition of gonadotrophin-releasing hormone. Evidence has accumulated indicating this to be due to a loss of the rat's ability to respond to markedly elevated estradiol with the usual arcuate nucleus neuro-glial plasticity that supports the estrogen-induced gonadotrophin surge (EIGS). Just as male rats are not capable of an EIGS, aged females loose this ability through repeated EIGS. Experiments indicate that in male rats the hypothalamic synaptology that develops as a result of exposure to testicular androgens in the perinatal period (brain sexual differentiation) is a result of conversion of testosterone from the testes to estrogen in the brain and is therefore due to early estrogen exposure. Aging females appear to reach a synaptology similar to males and constant estrus as a result of repeated exposure to ovarian estrogens during their reproductive careers. The relative role of aging and hormonal factors remains unclear. Morphological evidence is presented that indicates the above effects of estrogen involve changes in hypothalamic arcuate nucleus neurons and glia, including changes in the organization of perikaryal membranes as well as arcuate nucleus synaptology and the load of peroxidase in the astroglia. A possible role for free radicals (reactive oxygen species) in hypothalamic reproductive aging has been proposed. Such a mechanism is supported by evidence that the anti-oxidant vitamin E delays the onset of constant estrus and the accumulation of glial peroxidase in aging female rats. However, since the synaptology and peroxidase load in constant estrus females is independent of the age at which the constant estrus occurs, it appears that the role of (repeated) estradiol exposure is more deterministic of hypothalamic failure than is aging, per se.


Asunto(s)
Envejecimiento/fisiología , Estrógenos/fisiología , Hipotálamo/fisiología , Reproducción/fisiología , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Astrocitos/metabolismo , Ciclo Estral/fisiología , Femenino , Gonadotropinas Hipofisarias/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Masculino , Plasticidad Neuronal/fisiología , Ovario/fisiología , Peroxidasas/metabolismo , Ratas , Diferenciación Sexual/fisiología , Maduración Sexual/fisiología
14.
J Neuroendocrinol ; 14(6): 429-34, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12047717

RESUMEN

The effects of leptin on food intake, metabolism, sleep patterns and reproduction may be mediated, in part, by the midbrain serotonergic systems. Here, we report on the distribution of neurones that accumulate leptin in the raphe nuclei of male and female rats after intracerebroventricular administration of mouse recombinant leptin labelled with digoxigenin. Direct leptin-targeted cells were present in the periventricular grey, pontine and raphe nuclei. Confocal microscopy revealed that raphe neurones which accumulated leptin were predominantly serotonergic. The temporal pattern of leptin accumulation by raphe neurones showed a marked gender difference: 6 h after leptin administration, all male and female rats showed massive leptin binding in the dorsal raphe, while 30 min after leptin treatment, only 10% of male rats exhibited leptin-labelled cells in contrast to 50% of females. The present observations reveal that leptin can be selectively accumulated by serotonergic neurones in the raphe nuclei and that this mechanism is gender specific. These findings support the idea that the midbrain serotonergic system is an important mediator of the effects of leptin on brain function and may provide an explanation for gender differences in metabolism regulation and its coordination with higher functions of the brain.


Asunto(s)
Leptina/farmacocinética , Neuronas/metabolismo , Núcleos del Rafe/metabolismo , Serotonina/metabolismo , Animales , Femenino , Humanos , Inyecciones Intraventriculares , Leptina/administración & dosificación , Masculino , Ratones , Núcleos del Rafe/citología , Ratas , Ratas Wistar , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética , Caracteres Sexuales , Factores de Tiempo
15.
Brain Res Brain Res Rev ; 37(1-3): 25-37, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11744072

RESUMEN

This review summarizes the biosynthesis, cell type-distribution and function of brain aromatase cytochrome P450 (P450aro) and 5alpha-reductase enzymes. This overview covers the impact of the steroid products of the P450aro and 5alpha-reductase enzymes in establishing sexually dimorphic brain structures, specifically the sexually dimorphic nucleus of the preoptic area (SDN) and the anteroventral periventricular nucleus (AVPV). Additionally, since metabolites of the P450aro and 5alpha-reductase enzymes are known to regulate the calcium-binding protein, calbindin (CALB), CALB is reviewed in relationship to its potential role in determining sexually dimorphic brain structures. Finally, recent reports indicate that phytoestrogens inhibit P450aro and 5alpha-reductase activities in peripheral tissue sites, therefore, the effects of phytoestrogens on brain P450aro and 5alpha-reductase are briefly considered and the impact of consuming a high vs. a low phytoestrogen diet on visual spatial memory in male and female rats is presented.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/metabolismo , Andrógenos/metabolismo , Aromatasa/metabolismo , Encéfalo/enzimología , Isoflavonas , Progesterona/metabolismo , Inhibidores de 5-alfa-Reductasa , Animales , Inhibidores de la Aromatasa , Encéfalo/embriología , Calbindinas , Estrógenos no Esteroides/farmacología , Humanos , Fitoestrógenos , Preparaciones de Plantas , Proteína G de Unión al Calcio S100/metabolismo , Caracteres Sexuales
16.
Brain Res ; 919(1): 48-56, 2001 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-11689162

RESUMEN

Hypothalamic neuroendocrine dopaminergic neurons exhibit a diurnal rhythm. Higher level input to these neurons has not been described. In the present study, we identified fibers known to originate in the suprachiasmatic nucleus (SCN), which were associated with neuroendocrine dopaminergic neurons. Hypothalamic sections were obtained from either ovariectomized (OVX) female rats or OVX female rats implanted with estrogen and progesterone (E+P). Confocal microscopic images were acquired from the periventricular nucleus, as well as the rostral, dorsomedial, ventrolateral, and caudal regions of the arcuate nucleus. Using antibodies directed against vasoactive intestinal peptide (VIP) and tyrosine hydroxylase (TH) the rate-limiting enzyme in dopamine synthesis, fine VIP fibers in close apposition to TH-immunoreactive (IR) soma and proximal dendrites were revealed. Of the antibodies for the two VIP receptor subtypes (VIP1R and VIP2R), only VIP2R was found on TH-IR neurons. E+P significantly increased the incidence and density of neuroendocrine dopaminergic neurons expressing VIP2R, when compared to OVX animals. E+P did not affect the percent of neuroendocrine dopaminergic neurons associated with VIP fibers. No VIP fibers or VIP2R were found on dopaminergic neurons in the zona incerta. Brain sections triple labeled for Synapsin (a protein localized in synaptic vesicles) VIP, and TH demonstrated that Synapsin was colocalized with VIP fibers that were associated with TH-IR neurons in the arcuate nucleus. Double-label immuno-electron microscopy of hypothalamic sections labeled with antibodies for VIP and TH revealed VIP boutons associated with TH-IR soma and proximal dendrites. These results suggest VIPergic neurons may directly regulate neuroendocrine dopaminergic neuron activity, and ovarian steroids may play a modulatory role.


Asunto(s)
Dopamina/fisiología , Fibras Nerviosas/fisiología , Péptido Intestinal Vasoactivo/farmacología , Animales , Encéfalo/metabolismo , Encéfalo/fisiología , Dendritas/metabolismo , Dendritas/fisiología , Femenino , Inmunohistoquímica , Neuronas/metabolismo , Neuronas/fisiología , Sistemas Neurosecretores/metabolismo , Sistemas Neurosecretores/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Péptido Intestinal Vasoactivo/biosíntesis , Coloración y Etiquetado , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiología , Péptido Intestinal Vasoactivo/metabolismo
17.
Endocrinology ; 142(10): 4163-9, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11564668

RESUMEN

The recently discovered hormone, ghrelin, has been recognized as an important regulator of GH secretion and energy homeostasis. Orexigenic and adipogenic ghrelin is produced by the stomach, intestine, placenta, pituitary, and possibly in the hypothalamus. The concentration of circulating ghrelin, principally derived from the stomach, is influenced by acute and chronic changes in nutritional state. To date, most studies focused on the role of ghrelin in GH secretion or its function in complementing leptin action to prevent energy deficits. The potential significance of ghrelin in the etiology of obesity and cachexia as well as in the regulation of growth processes is the subject of ongoing discussions. A large quantity of information based on clinical trials and experimental studies with ghrelin and previously available synthetic ghrelin receptor agonists (GH secretagogues) must now be integrated with a rapidly increasing amount of data on the central regulation of metabolism and appetite. In this overview, we summarize recent findings and strategies on the integration of ghrelin into neuroendocrine networks that regulate energy homeostasis.


Asunto(s)
Hipotálamo/fisiología , Hormonas Peptídicas , Péptidos/fisiología , Animales , Metabolismo Energético/fisiología , Ghrelina , Humanos
18.
Exp Biol Med (Maywood) ; 226(8): 717-25, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11520936

RESUMEN

During amniote evolution, an early divergence occurred about 300 million years ago between the reptilian lines leading to the appearance of birds (anapsids) and mammals (synapsids). The different functional requirements of these vertebrate groups have involved divergent evolution of their brains. Even superficial examination reveals major anatomical differences between mammalian and avian brains, such as extensive development of the optic lobes and cerebellum in birds and a highly developed cortex in mammals. It has been nearly impossible to identify avian homologs of some mammalian brain regions by standard morphological criteria. This has long frustrated efforts at clarifying hypotheses regarding the anatomical location, field size, and regulation of brain functions shared between these two classes, despite the certainty that the principles of neurobiology apply equally at the cellular level in both groups. In an effort to remove this barrier, we have sought markers of common function that despite apparent anatomical dissimilarity, can allow recognition of homologous brain structures. We illustrate here how comparative analysis of the distribution of the steroid-metabolizing enzyme estrogen synthetase (aromatase) may help to understand the differences and similarities in the limbic system and hypothalamus of birds and mammals.


Asunto(s)
Aromatasa/biosíntesis , Hipotálamo/metabolismo , Sistema Límbico/metabolismo , Animales , Evolución Biológica , Aves , Chlorocebus aethiops , Coturnix , Humanos , Inmunohistoquímica , Ratas
19.
Nature ; 411(6836): 480-4, 2001 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-11373681

RESUMEN

The administration of leptin to leptin-deficient humans, and the analogous Lepob/Lepob mice, effectively reduces hyperphagia and obesity. But common obesity is associated with elevated leptin, which suggests that obese humans are resistant to this adipocyte hormone. In addition to regulating long-term energy balance, leptin also rapidly affects neuronal activity. Proopiomelanocortin (POMC) and neuropeptide-Y types of neurons in the arcuate nucleus of the hypothalamus are both principal sites of leptin receptor expression and the source of potent neuropeptide modulators, melanocortins and neuropeptide Y, which exert opposing effects on feeding and metabolism. These neurons are therefore ideal for characterizing leptin action and the mechanism of leptin resistance; however, their diffuse distribution makes them difficult to study. Here we report electrophysiological recordings on POMC neurons, which we identified by targeted expression of green fluorescent protein in transgenic mice. Leptin increases the frequency of action potentials in the anorexigenic POMC neurons by two mechanisms: depolarization through a nonspecific cation channel; and reduced inhibition by local orexigenic neuropeptide-Y/GABA (gamma-aminobutyric acid) neurons. Furthermore, we show that melanocortin peptides have an autoinhibitory effect on this circuit. On the basis of our results, we propose an integrated model of leptin action and neuronal architecture in the arcuate nucleus of the hypothalamus.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Leptina/fisiología , Red Nerviosa/fisiología , Neuronas/fisiología , Proopiomelanocortina/fisiología , Potenciales de Acción , Animales , Animales Modificados Genéticamente , Anorexia , Electrofisiología , Potenciales Evocados , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibición Neural , Neuropéptido Y/metabolismo , Ácido gamma-Aminobutírico/metabolismo
20.
Peptides ; 22(3): 473-81, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11287104

RESUMEN

Neuropeptide Y (NPY) stimulates and gamma-amino butyric acid (GABA) inhibits LH release in the rat. Since a sub-population of NPY-producing neurons in the arcuate nucleus (ARC) of the hypothalamus co-express GABA, the possibility of an interplay between NPY and GABA in the release of LH was investigated in two ways. First by employing light and electron microscopic double staining for NPY and GABA, using pre and post-immunolabeling on rat brain sections, we detected GABA in NPY immunoreactive axon terminals in the MPOA, one of the primary sites of action of these neurotransmitters/neuromodulators in the regulation of LH release. These morphological findings raised the possibility that inhibitory GABA co-released with NPY may act to restrain the excitatory effects of NPY on LH release. Muscimol (MUS, 0.44 or 1.76 nmol/rat), a GABA(A) receptor agonist, administered intracerebroventricularly (icv), alone failed to affect LH release, but NPY (0.47 nmol/rat icv) alone stimulated LH release in ovarian steroid-primed ovariectomized rats. On the other hand, administration of MUS blocked the NPY-induced stimulation of LH release in a dose-dependent manner. Similarly, administration of MUS abolished the excitatory effects on LH release of 1229U91, a selective NPY Y4 receptor agonist. These results support the possibility that in the event of co-release of these neurotransmitters/neuromodulators, GABA may act to restrain stimulation of LH release by NPY during the basal episodic and cyclic release of LH in vivo.


Asunto(s)
Hormona Luteinizante/metabolismo , Neuropéptido Y/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Encéfalo/embriología , Encéfalo/ultraestructura , Femenino , Inmunohistoquímica , Microscopía Electrónica , Microscopía Fluorescente , Modelos Biológicos , Muscimol/farmacología , Neuronas/metabolismo , Péptidos Cíclicos/farmacología , Unión Proteica , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...