Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Mol Psychiatry ; 23(5): 1157-1168, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28894301

RESUMEN

Increased motivation for highly rewarding food is a major contributing factor to obesity. Most of the literature focuses on the mesolimbic nuclei as the core of reward behavior regulation. However, the lateral hypothalamus (LH) is also a key reward-control locus in the brain. Here we hypothesize that manipulating glucagon-like peptide-1 receptor (GLP-1R) activity selectively in the LH can profoundly affect food reward behavior, ultimately leading to obesity. Progressive ratio operant responding for sucrose was examined in male and female rats, following GLP-1R activation and pharmacological or genetic GLP-1R blockade in the LH. Ingestive behavior and metabolic parameters, as well as molecular and efferent targets, of the LH GLP-1R activation were also evaluated. Food motivation was reduced by activation of LH GLP-1R. Conversely, acute pharmacological blockade of LH GLP-1R increased food motivation but only in male rats. GLP-1R activation also induced a robust reduction in food intake and body weight. Chronic knockdown of LH GLP-1R induced by intraparenchymal delivery of an adeno-associated virus-short hairpin RNA construct was sufficient to markedly and persistently elevate ingestive behavior and body weight and ultimately resulted in a doubling of fat mass in males and females. Interestingly, increased food reinforcement was again found only in males. Our data identify the LH GLP-1R as an indispensable element of normal food reinforcement, food intake and body weight regulation. These findings also show, for we believe the first time, that brain GLP-1R manipulation can result in a robust and chronic body weight gain. The broader implications of these findings are that the LH differs between females and males in its ability to control motivated and ingestive behaviors.


Asunto(s)
Conducta Alimentaria/fisiología , Receptor del Péptido 1 Similar al Glucagón/fisiología , Área Hipotalámica Lateral/metabolismo , Animales , Peso Corporal , Condicionamiento Operante/efectos de los fármacos , Dieta Alta en Grasa , Ingestión de Alimentos/efectos de los fármacos , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hipotálamo/metabolismo , Masculino , Motivación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Refuerzo en Psicología , Recompensa
2.
Psychoneuroendocrinology ; 119: 104720, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32563174

RESUMEN

Anxiety disorders are among the most prevalent categories of mental illnesses. The gut-brain axis, along with gastrointestinally-derived neuropeptides, like glucagon-like peptide-1 (GLP-1), are emerging as potential key regulators of emotionality, including anxiety behavior. However, the neuroanatomical substrates from which GLP-1 exerts its anxiogenic effect remain poorly characterized. Here we focus on a relatively new candidate nucleus, the supramammillary nucleus (SuM), located just caudal to the lateral hypothalamus and ventral to the ventral tegmental area. Our focus on the SuM is supported by previous data showing expression of GLP-1R mRNA throughout the SuM and activation of the SuM during anxiety-inducing behaviors in rodents. Data show that chemogenetic activation of neurons in the SuM results in an anxiolytic response in male and female rats. In contrast, selective activation of SuM GLP-1R, by microinjection of a GLP-1R agonist exendin-4 into the SuM resulted in potent anxiety-like behavior, measured in both open field and elevated plus maze tests in male and female rats. This anxiogenic effect of GLP-1R activation persisted after high-fat diet exposure. Importantly, reduction of GLP-1R expression in the SuM, by AAV-shRNA GLP-1R knockdown, resulted in a clear anxiolytic response; an effect only observed in female rats. Our data identify a new neural substrate for GLP-1 control of anxiety-like behavior and indicate that the SuM GLP-1R are sufficient for anxiogenesis in both sexes, but necessary only in females.


Asunto(s)
Ansiedad/psicología , Receptor del Péptido 1 Similar al Glucagón/fisiología , Hipotálamo Posterior/fisiología , Animales , Ansiedad/genética , Ansiedad/fisiopatología , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Exenatida/farmacología , Femenino , Técnicas de Silenciamiento del Gen , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor del Péptido 1 Similar al Glucagón/genética , Hipotálamo Posterior/efectos de los fármacos , Masculino , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas
3.
Neuroscience ; 180: 129-37, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21335062

RESUMEN

Ghrelin, a circulating orexigenic stomach-derived hormone, has recently been implicated in extra-homeostatic feeding, increasing food reward and food-motivated behavior. The precise target site(s) for ghrelin's effects on food reward have yet to be elucidated. The neurocircuitry underpinning food-motivated behavior involves, in particular, the dopamine cells of the ventral tegmental area (VTA) that project to the nucleus accumbens (NAcc). Ghrelin stimulation in both of these mesolimbic reward areas increases chow intake. Here we sought to determine if ghrelin acts directly within these mesolimbic reward areas to increase food reward/motivation in studies that combine feeding behavior, pharmacology, and neuroanatomy. We found that motivated behavior for a sucrose reward, assessed in an operant conditioning paradigm in rats, was increased when ghrelin was microinjected directly into the VTA but not into the NAcc. By contrast, ghrelin administration to both areas increased the free feeding of chow. Importantly, in a state of overnight food restriction, where endogenous levels of ghrelin are increased, ghrelin receptor (GHS-R1A) blockade in the VTA was sufficient to decrease the motivation to work for a sugar reward. Blockade of the GHS-R1A in VTA or NAcc was not sufficient to reduce fasting-induced chow hyperphagia. Taken together our data identify the VTA but not the NAcc as a direct, necessary, and sufficient target site for ghrelin's action on food motivation.


Asunto(s)
Conducta Alimentaria/fisiología , Ghrelina/metabolismo , Motivación/fisiología , Área Tegmental Ventral/metabolismo , Animales , Condicionamiento Operante , Alimentos , Ghrelina/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Área Tegmental Ventral/efectos de los fármacos
4.
Neuroscience ; 171(4): 1180-6, 2010 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-20933579

RESUMEN

Here we sought to determine whether ghrelin's central effects on food intake can be interrupted by nicotine acetylcholine receptor (nAChR) blockade. Ghrelin regulates mesolimbic dopamine neurons projecting from the ventral tegmental area (VTA) to the nucleus accumbens, partly via cholinergic VTA afferents originating in the laterodorsal tegmental area (LDTg). Given that these cholinergic projections to the VTA have been implicated in natural as well as drug-induced reinforcement, we sought to investigate the role of cholinergic signaling in ghrelin-induced food intake as well as fasting-induced food intake, for which endogenous ghrelin has been implicated. We found that i.p. treatment with the non-selective centrally active nAChR antagonist, mecamylamine decreased fasting-induced food intake in both mice and rats. Moreover, central administration of mecamylamine decreased fasting-induced food intake in rats. I.c.v. ghrelin-induced food intake was suppressed by mecamylamine i.p. but not by hexamethonium i.p., a peripheral nAChR antagonist. Furthermore, mecamylamine i.p. blocked food intake following ghrelin injection into the VTA. Expression of the ghrelin receptor, the growth hormone secretagogue receptor 1A, was found to co-localize with choline acetyltransferase, a marker of cholinergic neurons, in the LDTg. Finally, mecamylamine treatment i.p. decreased the ability of palatable food to condition a place preference. These data suggest that ghrelin-induced food intake is partly mediated via nAChRs and that nicotinic blockade decreases the rewarding properties of food.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Ghrelina/farmacología , Receptores Nicotínicos/fisiología , Transducción de Señal/fisiología , Área Tegmental Ventral/efectos de los fármacos , Análisis de Varianza , Animales , Colina O-Acetiltransferasa/metabolismo , Condicionamiento Operante/efectos de los fármacos , Vías de Administración de Medicamentos , Interacciones Farmacológicas , Ingestión de Alimentos/fisiología , Ayuno/fisiología , Preferencias Alimentarias/efectos de los fármacos , Preferencias Alimentarias/fisiología , Hexametonio/farmacología , Masculino , Mecamilamina/farmacología , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Antagonistas Nicotínicos/farmacología , Núcleo Accumbens/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Ghrelina/deficiencia , Receptores Nicotínicos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Área Tegmental Ventral/citología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda