Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Curr Psychiatry Rep ; 21(9): 85, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31410716

RESUMEN

PURPOSE OF REVIEW: This review synthesized the literature on predictors and mechanisms of post-bariatric alcohol problems, in order to guide future research on prevention and treatment targets. RECENT FINDINGS: Consistent evidence suggests an elevated risk of developing problems with alcohol following bariatric surgery. While there is a paucity of empirical data on predictors of problematic alcohol use after bariatric surgery, being male, a younger age, smoking, regular alcohol consumption, pre-surgical alcohol use disorder, and a lower sense of belonging have predicted alcohol misuse post-operatively. This review synthesizes potential mechanisms including specific bariatric surgical procedures, peptides and reinforcement/reward pathways, pharmacokinetics, and genetic influences. Finally, potential misperceptions regarding mechanisms are explored. Certain bariatric procedures elevate the risk of alcohol misuse post-operatively. Future research should serve to elucidate the complexities of reward signaling, genetically mediated mechanisms, and pharmacokinetics in relation to alcohol use across gender and developmental period by surgery type.


Asunto(s)
Consumo de Bebidas Alcohólicas/epidemiología , Alcoholismo/epidemiología , Cirugía Bariátrica/psicología , Obesidad Mórbida/psicología , Obesidad Mórbida/cirugía , Alcoholismo/complicaciones , Derivación Gástrica/psicología , Humanos , Obesidad Mórbida/complicaciones , Factores de Riesgo
2.
Am J Physiol Regul Integr Comp Physiol ; 313(5): R535-R548, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28768657

RESUMEN

Apolipoprotein AIV (ApoAIV) and cholecystokinin (CCK) are well-known satiating signals that are stimulated by fat consumption. Peripheral ApoAIV and CCK interact to prolong satiating signals. In the present study, we hypothesized that ApoAIV and CCK control energy homeostasis in response to high-fat diet feeding. To test this hypothesis, energy homeostasis in ApoAIV and CCK double knockout (ApoAIV/CCK-KO), ApoAIV knockout (ApoAIV-KO), and CCK knockout (CCK-KO) mice were monitored. When animals were maintained on a low-fat diet, ApoAIV/CCK-KO, ApoAIV-KO, and CCK-KO mice had comparable energy intake and expenditure, body weight, fat mass, fat absorption, and plasma parameters relative to the controls. In contrast, these KO mice exhibited impaired lipid transport to epididymal fat pads in response to intraduodenal infusion of dietary lipids. Furthermore, ApoAIV-KO mice had upregulated levels of CCK receptor 2 (CCK2R) in the small intestine while ApoAIV/CCK-KO mice had upregulated levels of CCK2R in the brown adipose tissue. After 20 wk of a high-fat diet, ApoAIV-KO and CCK-KO mice had comparable body weight and fat mass, as well as lower energy expenditure at some time points. However, ApoAIV/CCK-KO mice exhibited reduced body weight and adiposity relative to wild-type mice, despite having normal food intake. Furthermore, ApoAIV/CCK-KO mice displayed normal fat absorption and locomotor activity, as well as enhanced energy expenditure. These observations suggest that mice lacking ApoAIV and CCK have reduced body weight and adiposity, possibly due to impaired lipid transport and elevated energy expenditure.


Asunto(s)
Apolipoproteínas A/metabolismo , Colecistoquinina/metabolismo , Grasas de la Dieta/metabolismo , Homeostasis/fisiología , Adiposidad/genética , Adiposidad/fisiología , Animales , Apolipoproteínas A/deficiencia , Peso Corporal/fisiología , Colecistoquinina/deficiencia , Dieta con Restricción de Grasas/métodos , Ingestión de Alimentos/fisiología , Ingestión de Energía/fisiología , Metabolismo Energético/genética , Ratones Noqueados
3.
J Pediatr ; 167(5): 1042-8.e1, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26363548

RESUMEN

OBJECTIVE: To test the hypothesis that insulin secretion and insulin sensitivity would be improved in adolescents after Roux-en-Y gastric bypass (RYGB). STUDY DESIGN: A longitudinal study of 22 adolescents and young adults without diabetes undergoing laparoscopic RYGB (mean age 17.1 ± 1.42 years; range 14.5-20.1; male/female 8/14; Non-Hispanic White/African American 17/5) was conducted. Intravenous glucose tolerance tests were done to obtain insulin sensitivity (insulin sensitivity index), insulin secretion (acute insulin response to glucose ), and the disposition index as primary outcome variables. These variables were compared over the 1 year of observation using linear mixed modeling. RESULTS: In the 1-year following surgery, body mass index fell by 38% from a mean of 61 ± 12.3 to 39 ± 8.0 kg/m(2) (P < .01). Over the year following surgery, fasting glucose and insulin values declined by 54% and 63%, respectively. Insulin sensitivity index increased 300% (P < .01), acute insulin response to glucose decreased 56% (P < .01), leading to a nearly 2-fold increase in the disposition index (P < .01). Consistent with improved ß-cell function, the proinsulin to C-peptide ratio decreased by 21% (P < .01). CONCLUSIONS: RYGB reduced body mass index and improved both insulin sensitivity and ß-cell function in severely obese teens and young adults. These findings demonstrate that RYGB is associated with marked metabolic improvements in obese young people even as significant obesity persists. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00360373.


Asunto(s)
Derivación Gástrica , Resistencia a la Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Obesidad Infantil/metabolismo , Obesidad Infantil/cirugía , Adolescente , Glucemia/análisis , Índice de Masa Corporal , Ayuno , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Estudios Longitudinales , Masculino , Estudios Prospectivos , Resultado del Tratamiento , Adulto Joven
4.
Stress ; 17(6): 484-93, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25238021

RESUMEN

Behavioral modifications for the treatment of obesity, including caloric restriction, have notoriously low long-term success rates relative to bariatric weight-loss surgery. The reasons for the difference in sustained weight loss are not clear. One possibility is that caloric restriction alone activates the stress-responsive hypothalamo-pituitary-adrenocortical (HPA) axis, undermining the long-term maintenance of weight loss, and that this is abrogated after bariatric surgery. Accordingly, we compared the HPA response to weight loss in five groups of male rats: (1) high-fat diet-induced obese (DIO) rats treated with Roux-en-Y gastric bypass surgery (RYGB, n = 7), (2) DIO rats treated with vertical sleeve gastrectomy (VSG, n = 11), (3) DIO rats given sham surgery and subsequently restricted to the food intake of the VSG/RYGB groups (Pair-fed, n = 11), (4) ad libitum-fed DIO rats given sham surgery (Obese, n = 11) and (5) ad libitum chow-fed rats given sham surgery (Lean, n = 12). Compared with Lean controls, food-restricted rats exhibited elevated morning (nadir) non-stress plasma corticosterone concentration and increased hypothalamic corticotropin-releasing hormone and vasopressin mRNA expression, indicative of basal HPA activation. This was largely prevented when weight loss was achieved by bariatric surgery. DIO increased HPA activation by acute (novel environment) stress and this was diminished by bariatric surgery-, but not pair-feeding-, induced weight loss. These results indicate that the HPA axis is differentially affected by weight loss from caloric restriction versus bariatric surgery, and this may contribute to the differing long-term effectiveness of these two weight-loss approaches.


Asunto(s)
Restricción Calórica , Gastrectomía , Derivación Gástrica , Sistema Hipotálamo-Hipofisario/fisiopatología , Obesidad/dietoterapia , Obesidad/cirugía , Sistema Hipófiso-Suprarrenal/fisiopatología , Pérdida de Peso , Animales , Corticosterona/sangre , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Obesidad/sangre , Obesidad/etiología , Obesidad/fisiopatología , Sistema Hipófiso-Suprarrenal/metabolismo , ARN Mensajero/metabolismo , Ratas Long-Evans , Estrés Fisiológico , Factores de Tiempo , Vasopresinas/genética , Vasopresinas/metabolismo
5.
Appetite ; 61(1): 19-29, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23023044

RESUMEN

We have previously reported that a moderately high fat diet increases motivation for sucrose in adult rats. In this study, we tested the motivational, neurochemical, and metabolic effects of the high fat diet in male rats transitioning through puberty, during 5-8 weeks of age. We observed that the high fat diet increased motivated responding for sucrose, which was independent of either metabolic changes or changes in catecholamine neurotransmitter metabolites in the nucleus accumbens. However, AGRP mRNA levels in the hypothalamus were significantly elevated. We demonstrated that increased activation of AGRP neurons is associated with motivated behavior, and that exogenous (third cerebroventricular) AGRP administration resulted in significantly increased motivation for sucrose. These observations suggest that increased expression and activity of AGRP in the medial hypothalamus may underlie the increased responding for sucrose caused by the high fat diet intervention. Finally, we compared motivation for sucrose in pubertal vs. adult rats and observed increased motivation for sucrose in the pubertal rats, which is consistent with previous reports that young animals and humans have an increased preference for sweet taste, compared with adults. Together, our studies suggest that background diet plays a strong modulatory role in motivation for sweet taste in adolescent animals.


Asunto(s)
Dieta Alta en Grasa , Grasas de la Dieta/administración & dosificación , Sacarosa/administración & dosificación , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Composición Corporal , Cromatografía Líquida de Alta Presión , Ayuno , Prueba de Tolerancia a la Glucosa , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inmunohistoquímica , Masculino , Motivación/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
6.
Pharmacol Res ; 66(4): 283-91, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22750665

RESUMEN

Psychiatric patients frequently exhibit long-chain n-3 (LCn-3) fatty acid deficits and elevated triglyceride (TAG) production following chronic exposure to second generation antipsychotics (SGAs). Emerging evidence suggests that SGAs and LCn-3 fatty acids have opposing effects on stearoyl-CoA desaturase-1 (SCD1), which plays a pivotal role in TAG biosynthesis. Here we evaluated whether low LCn-3 fatty acid status would augment elevations in rat liver and plasma TAG concentrations following chronic treatment with the SGA risperidone (RSP), and evaluated relationships with hepatic SCD1 expression and activity indices. In rats maintained on the n-3 fatty acid-fortified (control) diet, chronic RSP treatment significantly increased liver SCD1 mRNA and activity indices (18:1/18:0 and 16:1/16:0 ratios), and significantly increased liver, but not plasma, TAG concentrations. Rats maintained on the n-3 deficient diet exhibited significantly lower liver and erythrocyte LCn-3 fatty acid levels, and associated elevations in LCn-6/LCn-3 ratio. In n-3 deficient rats, RSP-induced elevations in liver SCD1 mRNA and activity indices (18:1/18:0 and 16:1/16:0 ratios) and liver and plasma TAG concentrations were significantly greater than those observed in RSP-treated controls. Plasma glucose levels were not altered by diet or RSP, and body weight was lower in RSP- and VEH-treated n-3 deficient rats. These preclinical data support the hypothesis that low n-3 fatty acid status exacerbates RSP-induced hepatic steatosis by augmenting SCD1 expression and activity.


Asunto(s)
Antipsicóticos/efectos adversos , Ácidos Grasos Omega-3/metabolismo , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Hígado/metabolismo , Risperidona/efectos adversos , Estearoil-CoA Desaturasa/metabolismo , Animales , Glucemia/análisis , Dieta , Ingestión de Alimentos/efectos de los fármacos , Hígado Graso/patología , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , ARN Mensajero/genética , Ratas , Ratas Long-Evans , Estearoil-CoA Desaturasa/genética , Triglicéridos/sangre , Triglicéridos/metabolismo
7.
Nat Med ; 8(2): 179-83, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11821903

RESUMEN

Obesity and insulin resistance are major risk factors for a number of metabolic disorders, such as type 2 diabetes mellitus. Insulin has been suggested to function as one of the adiposity signals to the brain for modulation of energy balance. Administration of insulin into the brain reduces food intake and body weight, and mice with a genetic deletion of neuronal insulin receptors are hyperphagic and obese. However, insulin is also an anabolic factor; when administered systemically, pharmacological levels of insulin are associated with body weight gain in patients. In this study, we investigated the efficacy and feasibility of small molecule insulin mimetic compounds to regulate key parameters of energy homeostasis. Central intracerebroventricular (i.c.v.) administration of an insulin mimetic resulted in a dose-dependent reduction of food intake and body weight in rats, and altered the expression of hypothalamic genes known to regulate food intake and body weight. Oral administration of a mimetic in a mouse model of high-fat diet-induced obesity reduced body weight gain, adiposity and insulin resistance. Thus, insulin mimetics have a unique advantage over insulin in the control of body weight and hold potential as a novel anti-obesity treatment.


Asunto(s)
Reacción de Prevención/efectos de los fármacos , Benzoquinonas/farmacología , Peso Corporal/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Insulina/farmacología , Obesidad/prevención & control , Gusto/efectos de los fármacos , Animales , Apetito/efectos de los fármacos , Ventrículos Cerebrales/efectos de los fármacos , Ventrículos Cerebrales/fisiología , Dieta , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Intraventriculares , Resistencia a la Insulina , Masculino , Reacción en Cadena de la Polimerasa , Ratas , Ratas Endogámicas , Ratas Long-Evans , Sodio en la Dieta
8.
J Clin Invest ; 117(11): 3475-88, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17885689

RESUMEN

Disruptions of the melanocortin signaling system have been linked to obesity. We investigated a possible role of the central nervous melanocortin system (CNS-Mcr) in the control of adiposity through effects on nutrient partitioning and cellular lipid metabolism independent of nutrient intake. We report that pharmacological inhibition of melanocortin receptors (Mcr) in rats and genetic disruption of Mc4r in mice directly and potently promoted lipid uptake, triglyceride synthesis, and fat accumulation in white adipose tissue (WAT), while increased CNS-Mcr signaling triggered lipid mobilization. These effects were independent of food intake and preceded changes in adiposity. In addition, decreased CNS-Mcr signaling promoted increased insulin sensitivity and glucose uptake in WAT while decreasing glucose utilization in muscle and brown adipose tissue. Such CNS control of peripheral nutrient partitioning depended on sympathetic nervous system function and was enhanced by synergistic effects on liver triglyceride synthesis. Our findings offer an explanation for enhanced adiposity resulting from decreased melanocortin signaling, even in the absence of hyperphagia, and are consistent with feeding-independent changes in substrate utilization as reflected by respiratory quotient, which is increased with chronic Mcr blockade in rodents and in humans with loss-of-function mutations in MC4R. We also reveal molecular underpinnings for direct control of the CNS-Mcr over lipid metabolism. These results suggest ways to design more efficient pharmacological methods for controlling adiposity.


Asunto(s)
Sistema Nervioso Central/metabolismo , Metabolismo de los Lípidos , Melanocortinas/metabolismo , Transducción de Señal/fisiología , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Conducta Animal/fisiología , Ingestión de Alimentos , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Hormonas Estimuladoras de los Melanocitos/administración & dosificación , Hormonas Estimuladoras de los Melanocitos/metabolismo , Ratones , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Receptores de Melanocortina , alfa-MSH/administración & dosificación , alfa-MSH/análogos & derivados , alfa-MSH/metabolismo
9.
Nat Neurosci ; 9(3): 381-8, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16491079

RESUMEN

The gut hormone and neuropeptide ghrelin affects energy balance and growth hormone release through hypothalamic action that involves synaptic plasticity in the melanocortin system. Ghrelin binding is also present in other brain areas, including the telencephalon, where its function remains elusive. Here we report that circulating ghrelin enters the hippocampus and binds to neurons of the hippocampal formation, where it promotes dendritic spine synapse formation and generation of long-term potentiation. These ghrelin-induced synaptic changes are paralleled by enhanced spatial learning and memory. Targeted disruption of the gene that encodes ghrelin resulted in decreased numbers of spine synapses in the CA1 region and impaired performance of mice in behavioral memory testing, both of which were rapidly reversed by ghrelin administration. Our observations reveal an endogenous function of ghrelin that links metabolic control with higher brain functions and suggest novel therapeutic strategies to enhance learning and memory processes.


Asunto(s)
Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Memoria/fisiología , Hormonas Peptídicas/genética , Sinapsis/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/ultraestructura , Ghrelina , Hipocampo/efectos de los fármacos , Hipocampo/ultraestructura , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nootrópicos/metabolismo , Nootrópicos/farmacología , Hormonas Peptídicas/farmacología , Ratas , Ratas Sprague-Dawley , Percepción Espacial/efectos de los fármacos , Percepción Espacial/fisiología , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética
10.
Hippocampus ; 19(3): 235-52, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18831000

RESUMEN

The effects of selective ibotenate lesions of the complete hippocampus (CHip), the hippocampal ventral pole (VP), or the medial prefrontal cortex (mPFC) in male rats were assessed on several measures related to energy regulation (i.e., body weight gain, food intake, body adiposity, metabolic activity, general behavioral activity, conditioned appetitive responding). The testing conditions were designed to minimize the nonspecific debilitating effects of these surgeries on intake and body weight. Rats with CHip and VP lesions exhibited significantly greater weight gain and food intake compared with controls. Furthermore, CHip-lesioned rats, but not rats with VP lesions, showed elevated metabolic activity, general activity in the dark phase of the light-dark cycle, and greater conditioned appetitive behavior, compared with control rats without these brain lesions. In contrast, rats with mPFC lesions were not different from controls on any of these measures. These results indicate that hippocampal damage interferes with energy and body weight regulation, perhaps by disrupting higher-order learning and memory processes that contribute to the control of appetitive and consummatory behavior.


Asunto(s)
Conducta Apetitiva , Peso Corporal/fisiología , Conducta Alimentaria , Hipocampo/fisiología , Actividad Motora/fisiología , Corteza Prefrontal/fisiología , Adiposidad/fisiología , Análisis de Varianza , Animales , Composición Corporal , Calorimetría Indirecta , Ritmo Circadiano , Condicionamiento Psicológico , Metabolismo Energético , Alimentos , Masculino , Fotomicrografía , Ratas , Ratas Sprague-Dawley
11.
Physiol Behav ; 96(4-5): 593-601, 2009 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-19162054

RESUMEN

The current study sought to determine whether prenatal 3,4-methylenedioxy-N-methamphetamine (MDMA) exposure from E14-20 in the rat resulted in behavioral sequelae in adult offspring. Prenatal MDMA exposure results in increased dopaminergic fiber density in the prefrontal cortex, striatum and nucleus accumbens of young rats. Since these areas are critical in response to novelty, reward, attention and locomotor activity, we hypothesized that prenatal MDMA exposure would produce significant changes in the performance of tasks that examine such behaviors in adult rats. Adult rats prenatally exposed to MDMA exhibited greater activity and spent more time in the center during a novel open field test as compared to controls. This increased activity was not reflected in normal home cage activity. Prenatal exposure to MDMA did not affect feeding or food reward. It did not alter cocaine self-administration behaviors, nor did it have an effect on the locomotor response to amphetamine challenge. Finally, while prenatal MDMA did not affect performance in the radial arm maze or the Morris water maze (MWM), these animals demonstrated altered performance in a cued MWM paradigm. Prenatal MDMA exposure resulted in perseverative attendance to a hanging cue when the platform in the MWM was removed as compared to controls. Together, these data demonstrate that prenatal exposure to MDMA results in a behavioral phenotype in adult rats characterized by reduced anxiety, a heightened response to novelty, and "hyperattentiveness" to environmental cues during spatial learning.


Asunto(s)
Conducta Animal/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , N-Metil-3,4-metilenodioxianfetamina/toxicidad , Efectos Tardíos de la Exposición Prenatal , Serotoninérgicos/toxicidad , Conducta Espacial/efectos de los fármacos , Análisis de Varianza , Animales , Aprendizaje por Asociación/efectos de los fármacos , Atención/efectos de los fármacos , Cocaína/farmacología , Período Crítico Psicológico , Inhibidores de Captación de Dopamina/farmacología , Femenino , Edad Gestacional , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria a Corto Plazo , Actividad Motora/efectos de los fármacos , Embarazo , Distribución Aleatoria , Ratas , Autoadministración , Estadísticas no Paramétricas
12.
Int J Biochem Cell Biol ; 40(1): 28-45, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17698399

RESUMEN

Body weight is tightly regulated by a feedback mechanism involving peripheral adiposity signals and multiple central nervous system neurotransmitter pathways. Despite the tight regulation of body weight there is an increase in the prevalence of obesity and overweight in Western society. Obesity and overweight are conditions of excess body weight stored as fat. Syndecan-3, a member of the syndecan family of type I transmembrane heparan sulfate proteoglycans is a novel a regulator of feeding behavior and body weight. Syndecans are extracellular matrix molecules (ECMs) that modulate cell adhesion, cell-cell interactions and ligand-receptor interactions. The finding that syndecan-3 can regulate body weight is novel and provides a unique link between the extracellular matrix and body weight regulatory mechanisms. Uniquely, hormones such as leptin previously thought only to regulate body weight by modulating neuropeptide levels, have now been demonstrated to regulate neuronal plasticity in the hypothalamus. ECMs and syndecans have long been recognized as regulators of plasticity. Therefore, this review will focus on highlighting the role of syndecans and in particular syndecan-3 in neuronal development and synaptic organization and how these processes may integrate body weight regulation. As part of this review, we will highlight how syndecan-3 can mediate the activity of adiposity signals, such as leptin, and facilitate changes in neuronal plasticity.


Asunto(s)
Peso Corporal , Plasticidad Neuronal , Sindecanos , Animales , Peso Corporal/fisiología , Encéfalo/embriología , Encéfalo/fisiología , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Humanos , Leptina/genética , Leptina/metabolismo , Ratones , Ratones Noqueados , Vías Nerviosas/fisiología , Plasticidad Neuronal/fisiología , Obesidad/etiología , Obesidad/genética , Obesidad/metabolismo , Polimorfismo Genético , Ratas , Transmisión Sináptica , Sindecanos/genética , Sindecanos/metabolismo
13.
Endocrinology ; 149(12): 6416-24, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18687780

RESUMEN

Circulating angiotensin II (ANGII) elicits water intake and activates the hypothalamic-pituitary-adrenal (HPA) axis by stimulating angiotensin type 1 receptors (AT1Rs) within circumventricular organs. The subfornical organ (SFO) and the organum vasculosum of the lamina terminalis (OVLT) are circumventricular organs that express AT1Rs that bind blood-borne ANGII and stimulate integrative and effector regions of the brain. The goal of these studies was to determine the contribution of AT1Rs within the SFO and OVLT to the water intake and HPA response to increased circulating ANGII. Antisense oligonucleotides directed against the AT1R [AT1R antisense (AT1R AS)] were administered into the OVLT or SFO. Quantitative receptor autoradiography confirmed that AT1R AS decreased ANGII binding in the SFO and OVLT compared with the scrambled sequence control but did not affect AT1R binding in other nuclei. Subsequently, water intake, ACTH, and corticosterone (CORT) were assessed after administration of isoproterenol, a beta-adrenergic agonist that decreases blood pressure and elevates circulating ANGII. Delivery of AT1R AS into the SFO attenuated water intake, ACTH, and CORT after isoproterenol, whereas similar treatment in the OVLT had no effect. To determine the specificity of this blunted drinking and HPA response, the same parameters were measured after treatment with hypertonic saline, a stimulus that induces drinking independently of ANGII. Delivery of AT1R AS into the SFO or OVLT had no effect on water intake, ACTH, or CORT after hypertonic saline. The results imply that AT1R within the SFO mediate drinking and HPA responses to stimuli that increase circulating ANGII.


Asunto(s)
Ingestión de Líquidos/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Isoproterenol/farmacología , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo , Órgano Subfornical/efectos de los fármacos , Agonistas Adrenérgicos beta/farmacología , Hormona Adrenocorticotrópica/sangre , Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Corticosterona/sangre , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/fisiología , Masculino , Oligodesoxirribonucleótidos Antisentido/farmacología , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiología , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Receptor de Angiotensina Tipo 1/fisiología , Cloruro de Sodio/farmacología , Órgano Subfornical/metabolismo , Órgano Subfornical/fisiología
14.
Endocrinology ; 149(6): 3009-15, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18308842

RESUMEN

We recently reported that the hypothalamic homeobox domain transcription factor Bsx plays an essential role in the central nervous system control of spontaneous physical activity and the generation of hyperphagic responses. Moreover, we found Bsx to be a master regulator for the hypothalamic expression of key orexigenic neuropeptide Y and agouti gene-related protein. We now hypothesized that Bsx, which is expressed in the dorsomedial and arcuate nucleus (ARC) of the hypothalamus, is regulated by afferent signals in response to peripheral energy balance. Bsx expression was analyzed using in situ hybridization in fed vs. fasted (24 h) and ghrelin vs. leptin-treated rats, as well as in mice deficient for leptin or the ghrelin signaling. Ghrelin administration increased, whereas ghrelin receptor antagonist decreased ARC Bsx expression. Leptin injection attenuated the fasting-induced increase in ARC Bsx levels but had no effect in fed rats. Dorsomedial hypothalamic nucleus Bsx expression was unaffected by pharmacological modifications of leptin or ghrelin signaling. Obese leptin-deficient (ob/ob) mice, but not obese melanocortin 4 receptor-knockout mice, showed higher expression of Bsx, consistent with dependency from afferent leptin rather than increased adiposity per se. Interestingly, exposure to a high-fat diet triggered Bsx expression, consistent with the concept that decreased leptin signaling due to a high-fat diet induced leptin resistance. Our data indicate that ARC Bsx expression is specifically regulated by afferent energy balance signals, including input from leptin and ghrelin. Future studies will be necessary to test if Bsx may be involved in the pathogenesis of leptin resistance.


Asunto(s)
Ingestión de Alimentos/fisiología , Actividad Motora/fisiología , Proteína X Asociada a bcl-2/fisiología , Animales , Metabolismo Energético , Ayuno , Ghrelina/sangre , Insulina/sangre , Leptina/sangre , Masculino , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Proteína X Asociada a bcl-2/genética
15.
Diabetes ; 56(4): 1051-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17251274

RESUMEN

Ghrelin, the only known orexigenic gut hormone, is secreted mainly from the stomach, increases with fasting and before meal initiation in humans and rats, and increases food intake after central or peripheral administration. To investigate sex differences in the action of ghrelin, we assessed the effects of exogenous ghrelin in intact male and female rats, the effects of exogenous ghrelin in ovariectomized (OVX) and estradiol (E2)-treated female rats, as well as the effects of OVX on plasma ghrelin and hypothalamic orexigneic neuropeptide expression in rats and on food intake and weight gain in transgenic mice lacking the ghrelin receptor (Ghsr(-/-) mice). Male and OVX female rats were significantly more sensitive than intact female rats to the orexigenic effects of both centrally (intra-third ventricular, i3vt, 0.01, 0.1, and 1.0 nmol) and systemically (ip, 3, 6, and 9 nmol) administered ghrelin. This difference is likely to be estradiol dependent because E2 attenuated the orexigenic action of ghrelin in OVX female and male rats. Furthermore, OVX increased food intake and body weight in wild-type mice, but not in Ghsr(-/-) mice, suggesting that OVX increases food intake by releasing ghrelin from a tonic inhibitory effect of estradiol. In addition, following OVX, there was an increase in plasma ghrelin that was temporally associated with increased food intake, body weight, and hypothalamic neuropeptide Y and Agouti-related protein mRNA expression. Collectively, these data suggest that estradiol inhibits the orexigenic action of ghrelin in females, that weight gain associated with OVX is ghrelin mediated, and that this endocrine interaction may account for an important sex differences in food intake and the regulation of body weight.


Asunto(s)
Estradiol/farmacología , Conducta Alimentaria/fisiología , Ovariectomía , Hormonas Peptídicas/fisiología , Animales , Peso Corporal , Cartilla de ADN , Ingestión de Energía , Femenino , Ghrelina , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Hormonas Peptídicas/deficiencia , Hormonas Peptídicas/efectos de los fármacos , Hormonas Peptídicas/genética , Reacción en Cadena de la Polimerasa , Ratas , Ratas Long-Evans , Caracteres Sexuales
16.
J Pharmacol Exp Ther ; 324(1): 206-13, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17932246

RESUMEN

The melanin-concentrating hormone-1 receptor (MCH1R) is a G-protein-coupled receptor expressed in the brain and peripheral tissues that regulates energy storage and body weight. Here, we focused on discovery of the mechanism and site of action for a small-molecule MCH1R antagonist, which yields weight loss in a mouse model of human obesity. MCH1R is expressed throughout the brain but also found in peripheral tissues known to regulate fat storage and utilization, e.g., skeletal muscle and adipose tissue. Previous studies of MCH1R antagonist studies have not delineated the site that is critical for mediating the anorexigenic and weight-reducing actions. In this study, we evaluated the role of the brain and peripheral tissue receptors. We developed a novel nonbrain-permeable MCH antagonist analog with a carboxylic acid moiety to specifically test the site of action. Based on in vitro and in vivo assays, the analog is not able to cross the blood-brain barrier and does not lead to inhibition of food intake and reduced body weight. The data clearly demonstrate that MCH1R antagonists need access to the brain to reduce body weight and fat mass. The brain-permeable MCH1R antagonist leads to significant reduction in body weight and fat mass in diet-induced obese mice. The effect is dose-dependent and appears to be partially driven by a reduction in food intake. Finally, these studies show the utility of a medicinal chemistry approach to address an important biological and pharmacological question.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Encéfalo/metabolismo , Obesidad/tratamiento farmacológico , Receptores de Somatostatina/antagonistas & inhibidores , Animales , Fármacos Antiobesidad/metabolismo , Fármacos Antiobesidad/farmacocinética , Reacción de Prevención , Peso Corporal/efectos de los fármacos , Células CACO-2 , Línea Celular , Grasas de la Dieta/farmacología , Ingestión de Alimentos/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Long-Evans , Receptores de Somatostatina/metabolismo , Gusto
17.
Behav Neurosci ; 122(6): 1257-63, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19045945

RESUMEN

Recent studies indicate that decreased central dopamine is associated with diet-induced obesity in humans and in animal models. In the current study, the authors assessed the hypothesis that diet-induced obesity reduces mesolimbic dopamine function. Specifically, the authors compared dopamine turnover in this region between rats fed a high-fat diet and those consuming a standard low-fat diet. The authors also assessed behavioral consequences of diet-induced obesity by testing the response of these animals in a conditioned place paradigm using amphetamine as a reinforcer and in an operant conditioning paradigm using sucrose reinforcement. Results demonstrate that animals consuming a high-fat diet, independent of the development of obesity, exhibit decreased dopamine turnover in the mesolimbic system, reduced preference for an amphetamine cue, and attenuated operant responding for sucrose. The authors also observed that diet-induced obesity with a high-fat diet attenuated mesolimbic dopamine turnover in the nucleus accumbens. These data are consistent with recent hypotheses that the hormonal signals derived from adipose tissue regulate the activity of central nervous system structures involved in reward and motivation, which may have implications for the treatment of obesity and/or addiction.


Asunto(s)
Anfetamina/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Grasas de la Dieta/administración & dosificación , Dopamina/metabolismo , Sistema Límbico/efectos de los fármacos , Recompensa , Análisis de Varianza , Animales , Conducta Animal , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Sistema Límbico/metabolismo , Locomoción/efectos de los fármacos , Masculino , Ratas , Ratas Long-Evans , Esquema de Refuerzo , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación
18.
Peptides ; 29(1): 139-47, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18068269

RESUMEN

Recent conceptualizations of food intake have divided ingestive behavior into multiple distinct phases. Here, we present a temporally and operationally defined classification of ingestive behaviors. Importantly, various physiological signals including hypothalamic peptides are thought to impact these distinct behavioral phases of ingestion differently. In this review, we summarize a number of behavioral assays designed to delineate the effects of hormone and peptide signals that influence food intake on these ingestive mechanisms. Finally, we discuss two issues that we have encountered in our laboratory which may obstruct the interpretation of results from these types of studies. First, the influence of previous experience with foods used in these behavioral tests and second, the importance of the nutrient composition of the selected test foods. The important conclusion discussed here is that the behavioral analysis of ingestion is accompanied by theoretical constructs and artificial divisions of biological realities and the appreciation of this fact can only increase the opportunities of contemporary behavioral scientists to make significant and novel observations of ingestive behaviors.


Asunto(s)
Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Animales , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Hormonas/farmacología , Hormonas/fisiología , Aprendizaje/efectos de los fármacos , Motivación , Péptidos/farmacología , Péptidos/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
19.
Physiol Behav ; 93(4-5): 912-8, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18234242

RESUMEN

Previous studies have shown that administration of the fatty acids, linoleic and oleic acid, either by intragastric or intraintestinal infusion, suppresses food intake and body weight in rats. While still not fully understood, gut-mediated satiety mechanisms likely are potential effectors of this robust response to gastrointestinal fatty acid infusions. The objective of this study was to assess the effects of voluntary access to an oleic acid derivative, ethyl oleate (EO), on subsequent food intake and body weight in rats. Animals were randomized either to a 12.5% EO diet or a soybean oil diet as a "breakfast," followed either by two one-hour or one five-hour access periods to standard rodent diet, and food intake and body weights were collected. Across 14 days access, rats consuming EO on both feeding schedules gained less weight and consumed less total kilocalories than rats consuming the SO diet. Further, plasma levels of glucose and insulin were comparable in both EO and SO diet groups. In summary, EO was found to increase weight loss in rats maintained on a 75% food-restriction regimen, and attenuate weight-gain upon resumption of an ad-libitum feeding regimen. These data indicate that voluntary access to EO promoted short-term satiety, compared to SO diet, and that these effects contributed to an important and novel attenuated weight gain in EO-fed animals.


Asunto(s)
Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ácidos Oléicos/administración & dosificación , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Peso Corporal/fisiología , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/fisiología , Masculino , Ratas , Ratas Long-Evans , Factores de Tiempo
20.
Nutrition ; 24(9): 843-7, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18725081

RESUMEN

The regulation of energy balance depends on the precise co-ordination of multiple peripheral and central systems. Much recent research has highlighted the importance of behavioral mechanisms is this control and suggested that the regulation of body weight shares central nervous system pathways in common with other complex behaviors, including learning and drug addiction. We present a brief review of some of this work and highlight the novel functions for central orexigenic neuropeptides. We review evidence that organisms engage in critical regulatory behaviors before and after ingestion has occurred. Additional evidence supports the idea that appetitive mechanisms are engaged that are critical for the regulation of intake during the act of ingestion. We briefly discuss the recent work on the potential role for central nervous system reward centers, how those might be critically linked to the central regulation of food intake, and how they may be dysregulated by the abundance of highly palatable, energy-dense foods.


Asunto(s)
Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neuropéptido Y/genética , Neuropéptido Y/fisiología , Neuropéptidos/genética , Neuropéptidos/fisiología , Animales , Regulación del Apetito/genética , Regulación del Apetito/fisiología , Ingestión de Alimentos/genética , Ingestión de Alimentos/fisiología , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Humanos , Orexinas , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA