Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell Metab ; 5(5): 383-93, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17488640

RESUMEN

The importance of neuropeptides in the hypothalamus has been experimentally established. Due to difficulties in assessing function in vivo, the roles of the fast-acting neurotransmitters glutamate and GABA are largely unknown. Synaptic vesicular transporters (VGLUTs for glutamate and VGAT for GABA) are required for vesicular uptake and, consequently, synaptic release of neurotransmitters. Ventromedial hypothalamic (VMH) neurons are predominantly glutamatergic and express VGLUT2. To evaluate the role of glutamate release from VMH neurons, we generated mice lacking VGLUT2 selectively in SF1 neurons (a major subset of VMH neurons). These mice have hypoglycemia during fasting secondary to impaired fasting-induced increases in the glucose-raising pancreatic hormone glucagon and impaired induction in liver of mRNAs encoding PGC-1alpha and the gluconeogenic enzymes PEPCK and G6Pase. Similarly, these mice have defective counterregulatory responses to insulin-induced hypoglycemia and 2-deoxyglucose (an antimetabolite). Thus, glutamate release from VMH neurons is an important component of the neurocircuitry that functions to prevent hypoglycemia.


Asunto(s)
Ácido Glutámico/metabolismo , Hipoglucemia/metabolismo , Hipotálamo/citología , Neuronas/metabolismo , Sinapsis/metabolismo , Animales , Electrofisiología , Glucagón/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Hibridación in Situ , Insulina , Hígado/metabolismo , Ratones , Ratones Transgénicos , Neuronas/citología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transactivadores/metabolismo , Factores de Transcripción , Proteína 2 de Transporte Vesicular de Glutamato/genética
2.
Neuron ; 49(2): 191-203, 2006 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-16423694

RESUMEN

Leptin, an adipocyte-derived hormone, acts directly on the brain to control food intake and energy expenditure. An important question is the identity of first-order neurons initiating leptin's anti-obesity effects. A widely held view is that most, if not all, of leptin's effects are mediated by neurons located in the arcuate nucleus of the hypothalamus. However, leptin receptors (LEPRs) are expressed in other sites as well, including the ventromedial hypothalamus (VMH). The possible role of leptin acting in "nonarcuate" sites has largely been ignored. In the present study, we show that leptin depolarizes and increases the firing rate of steroidogenic factor-1 (SF1)-positive neurons in the VMH. We also show, by generating mice that lack LEPRs on SF1-positive neurons, that leptin action at this site plays an important role in reducing body weight and, of note, in resisting diet-induced obesity. These results reveal a critical role for leptin action on VMH neurons.


Asunto(s)
Peso Corporal/fisiología , Proteínas de Homeodominio/fisiología , Homeostasis/fisiología , Leptina/farmacología , Neuronas/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/fisiología , Animales , Composición Corporal/efectos de los fármacos , Composición Corporal/genética , Composición Corporal/fisiología , Dieta , Electrofisiología , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Homeostasis/efectos de los fármacos , Inmunohistoquímica , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Obesidad/fisiopatología , Técnicas de Placa-Clamp , Fenotipo , Sondas ARN , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Leptina , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor Esteroidogénico 1 , Factores de Transcripción/genética , Núcleo Hipotalámico Ventromedial/citología
3.
J Neurosci ; 29(47): 14828-35, 2009 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19940178

RESUMEN

The adipose-derived hormone, leptin, was discovered over 10 years ago, but only now are we unmasking its downstream pathways which lead to reduced energy intake (feeding) and increased energy expenditure (thermogenesis). Recent transgenic models have challenged the long-standing supposition that the hypothalamic arcuate nucleus (Arc) is omnipotent in the central response to leptin, and research focus is beginning to shift to examine roles of extra-arcuate sites. Dhillon et al. (2006) demonstrated that targeted knock out of the signaling form of the leptin receptor (lepr-B) in steroidogenic factor 1 (SF-1) cells of the hypothalamic ventromedial nucleus (VMN) produces obesity of a similar magnitude to the pro-opiomelanocortin (POMC)-driven lepr-B deleted mouse, via a functionally distinct mechanism. These findings reveal that SF-1 cells of the VMN could be equally as important as POMC cells in mediating leptin's anti-obesity effects. However, the identification of molecular and cellular correlates of this relationship remains tantalizingly unknown. Here, we have shown that mRNA expression of the VMN-expressed neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is regulated according to energy status and that it exerts catabolic effects when administered centrally to mice. Furthermore, we have shown that SF-1 and PACAP mRNAs are colocalized in the VMN, and that leptin signaling via lepr-B is required for normal PACAP expression in these cells. Finally, blocking endogenous central PACAP signaling with the antagonist PACAP(6-38) markedly attenuates leptin-induced hypophagia and hyperthermia in vivo. Thus, it appears that PACAP is an important mediator of central leptin effects on energy balance.


Asunto(s)
Leptina/metabolismo , Neuronas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Factor Esteroidogénico 1/metabolismo , Núcleo Hipotalámico Ventromedial/metabolismo , Animales , Regulación del Apetito/fisiología , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Fiebre/tratamiento farmacológico , Fiebre/metabolismo , Fiebre/fisiopatología , Leptina/farmacología , Masculino , Ratones , Neuronas/citología , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Receptores de Leptina/efectos de los fármacos , Receptores de Leptina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor Esteroidogénico 1/genética , Núcleo Hipotalámico Ventromedial/citología
4.
Endocrinology ; 149(11): 5654-61, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18669597

RESUMEN

Suppressor of cytokine signaling 3 (Socs3) has been identified as a mediator of central leptin resistance, but the identity of specific neurons in which Socs3 acts to suppress leptin signaling remains elusive. The ventromedial hypothalamus (VMH) was recently shown to be an important site for leptin action because deleting leptin receptor within VMH neurons causes obesity. To examine the role of VMH Socs3 in leptin resistance and energy homeostasis, we generated mice lacking Socs3 specifically in neurons positive for steroidogenic factor 1 (SF1), which is expressed abundantly in the VMH. These mice had increased phosphorylation of signal transducer and activator of transcription-3 in VMH neurons, suggesting improved leptin signaling, and consistently, food intake and weight-reducing effects of exogenous leptin were enhanced. Furthermore, on either chow or high-fat diets, these mice had reduced food intake. Unexpectedly, energy expenditure was reduced as well. Mice lacking Socs3 in SF1 neurons, despite no change in body weight, had improved glucose homeostasis and were partially protected from hyperglycemia and hyperinsulinemia induced by high-fat diets. These results suggest that Socs3 in SF1 neurons negatively regulates leptin signaling and plays important roles in mediating leptin sensitivity, glucose homeostasis, and energy expenditure.


Asunto(s)
Peso Corporal/genética , Glucosa/metabolismo , Neuronas/metabolismo , Factor Esteroidogénico 1/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Animales , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Homeostasis/genética , Insulina/sangre , Leptina/farmacología , Masculino , Ratones , Ratones Transgénicos , Especificidad de Órganos/genética , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Núcleo Hipotalámico Ventromedial/efectos de los fármacos , Núcleo Hipotalámico Ventromedial/metabolismo
5.
Diabetes ; 54(4): 1023-31, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15793240

RESUMEN

Local glucocorticoid (GC) action depends on intracellular GC metabolism by 11beta-hydroxysteroid dehydrogenases (11betaHSDs). 11betaHSD1 activates GCs, while 11betaHSD2 inactivates GCs. Adipocyte-specific amplification of GCs through transgenic overexpression of 11betaHSD1 produces visceral obesity and the metabolic syndrome in mice. To determine whether adipocyte-specific inactivation of GCs protects against this phenotype, we created a transgenic model in which human 11betaHSD2 is expressed under the control of the murine adipocyte fatty acid binding protein (aP2) promoter (aP2-h11betaHSD2). Transgenic mice have increased 11betaHSD2 expression and activity exclusively in adipose tissue, with the highest levels in subcutaneous adipose tissue, while systemic indexes of GC exposure are unchanged. Transgenic mice resist weight gain on high-fat diet due to reduced fat mass accumulation. This improved energy balance is associated with decreased food intake, increased energy expenditure, and improved glucose tolerance and insulin sensitivity. Adipose tissue gene expression in transgenic mice is characterized by decreased expression of leptin and resistin and increased expression of adiponectin, peroxisome proliferator-activated receptor gamma, and uncoupling protein 2. These data suggest that reduction of active GCs exclusively in adipose tissue is an important determinant of a favorable metabolic phenotype with respect to energy homeostasis and the metabolic syndrome.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Adipocitos/fisiología , Glucocorticoides/fisiología , Obesidad/fisiopatología , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Adipocitos/enzimología , Tejido Adiposo , Envejecimiento , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Grasas de la Dieta , Proteínas de Unión a Ácidos Grasos , Expresión Génica , Resistencia a la Insulina , Síndrome Metabólico/enzimología , Síndrome Metabólico/fisiopatología , Ratones , Ratones Transgénicos , Obesidad/enzimología , Regiones Promotoras Genéticas , Aumento de Peso
6.
Science ; 352(6281): 54-61, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26917594

RESUMEN

Defects in the mitochondrial respiratory chain (RC) underlie a spectrum of human conditions, ranging from devastating inborn errors of metabolism to aging. We performed a genome-wide Cas9-mediated screen to identify factors that are protective during RC inhibition. Our results highlight the hypoxia response, an endogenous program evolved to adapt to limited oxygen availability. Genetic or small-molecule activation of the hypoxia response is protective against mitochondrial toxicity in cultured cells and zebrafish models. Chronic hypoxia leads to a marked improvement in survival, body weight, body temperature, behavior, neuropathology, and disease biomarkers in a genetic mouse model of Leigh syndrome, the most common pediatric manifestation of mitochondrial disease. Further preclinical studies are required to assess whether hypoxic exposure can be developed into a safe and effective treatment for human diseases associated with mitochondrial dysfunction.


Asunto(s)
Enfermedad de Leigh/genética , Enfermedad de Leigh/terapia , Mitocondrias/metabolismo , Oxígeno/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Anaerobiosis , Animales , Antimicina A/análogos & derivados , Antimicina A/farmacología , Proteínas Bacterianas , Biomarcadores/sangre , Temperatura Corporal , Peso Corporal , Proteína 9 Asociada a CRISPR , Modelos Animales de Enfermedad , Transporte de Electrón/efectos de los fármacos , Complejo I de Transporte de Electrón/genética , Endonucleasas , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Técnicas de Inactivación de Genes , Estudio de Asociación del Genoma Completo , Glicina/análogos & derivados , Glicina/farmacología , Glicina/uso terapéutico , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Isoquinolinas/farmacología , Isoquinolinas/uso terapéutico , Células K562 , Enfermedad de Leigh/patología , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Respiración , Supresión Genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/antagonistas & inhibidores , Pez Cebra
7.
Diabetes ; 51(6): 1729-36, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12031959

RESUMEN

Recombinant adeno-associated virus (rAAV), encoding either rat leptin (rAAV-lep) or green fluorescent protein (rAAV-GFP, control), was injected intracerebroventricularly in rats consuming a high-fat diet (HFD; 45 kcal%). Caloric consumption and body weight were monitored weekly until the rats were killed at 9 weeks. Untreated control rats consuming regular rat diet (RCD; 11 kcal%) were monitored in parallel. Body weight gain was accelerated in rAAV-GFP + HFD control rats relative to those consuming RCD, despite equivalent kcal consumption. At 9 weeks, serum leptin, free fatty acids, triglycerides, and insulin were elevated in HFD control rats. In contrast, rAAV-lep treatment reduced intake and blocked the HFD-induced increase in weight, adiposity, and metabolic variables. Blood glucose was slightly reduced but within the normal range, and serum ghrelin levels were significantly elevated in rAAV-lep + HFD rats. Uncoupling protein-1 (UCP1) mRNA in brown adipose tissue (BAT), an index of energy expenditure through nonshivering thermogenesis, was decreased in rats consuming HFD. Treatment with rAAV-lep significantly augmented BAT UCP1 mRNA expression, indicating increased thermogenic energy expenditure. These findings demonstrate that central leptin gene therapy efficiently prevents weight gain, increased adiposity, and hyperinsulinemia in rats consuming an HFD by decreasing energy intake and increasing thermogenic energy expenditure.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Terapia Genética , Hiperinsulinismo/prevención & control , Leptina/genética , Hormonas Peptídicas , Péptidos/sangre , Aumento de Peso , Tejido Adiposo , Tejido Adiposo Pardo/química , Animales , Composición Corporal , Proteínas Portadoras/genética , Dependovirus/genética , Ingestión de Energía , Metabolismo Energético , Ácidos Grasos no Esterificados/sangre , Técnicas de Transferencia de Gen , Vectores Genéticos , Ghrelina , Proteínas Fluorescentes Verdes , Canales Iónicos , Leptina/sangre , Proteínas Luminiscentes/genética , Masculino , Proteínas de la Membrana/genética , Proteínas Mitocondriales , Obesidad/prevención & control , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Termogénesis , Triglicéridos/sangre , Proteína Desacopladora 1
8.
Endocrinology ; 145(6): 2767-74, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15016719

RESUMEN

Hyperthyroidism and states of adrenergic hyperactivity have many common clinical features, suggesting similar pathogenic mechanisms of action. The widespread use of beta-adrenergic receptor (betaAR) antagonists (beta-blockers) to treat hyperthyroidism has led to the belief that the physiological consequences of thyroid hormone (TH) excess are mediated in part via catecholamine signaling through betaARs. To test this hypothesis, we compared the response to TH excess in mice lacking the three known betaARs (beta-less) vs. wild-type (WT) mice. Although beta-less mice had a lower heart rate at baseline in comparison to WT mice, the metabolic and cardiovascular responses to hyperthyroidism were equivalent in both WT and beta-less mice. These data indicate that the metabolic and cardiovascular effects of TH excess are largely independent of betaARs. These findings suggest that the efficacy of clinical treatment of hyperthyroidism with beta-blockers is due to antagonism of sympathetic signaling, and that this process functions independently of TH action.


Asunto(s)
Sistema Cardiovascular/fisiopatología , Hipertiroidismo/fisiopatología , Receptores Adrenérgicos beta/metabolismo , Agonistas Adrenérgicos beta/farmacología , Animales , Hipertiroidismo/inducido químicamente , Hipertiroidismo/metabolismo , Isoproterenol/farmacología , Ratones , Receptores Adrenérgicos beta/deficiencia , Triyodotironina
9.
Peptides ; 23(5): 975-84, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12084530

RESUMEN

Leukemia inhibitory factor (LIF) overexpression, induced by the intracerebroventricular (i.c.v.) injection of an recombinant adeno-associated viral vector encoding LIF (rAAV-LIF), resulted in a dose-dependent reduction in body weight (BW) gain, food intake (FI) and adiposity, evidenced by suppression of serum leptin and free fatty acids for an extended period in outbred adult female rats. A dose-dependent reduction in serum insulin levels and unchanged serum glucose, energy expenditure through thermogenesis as indicated by uncoupling protein-1 (UCP-1) mRNA expression in brown adipose tissue (BAT), and metabolism as indicated by serum T3 and T4, accompanied the blockade of weight gain. Thus, central rAAV-LIF therapy is a viable strategy to voluntarily reduce appetite and circumvent leptin resistance, a primary factor underlying age-dependent weight gain and obesity in rodents and humans.


Asunto(s)
Apetito/genética , Peso Corporal/genética , Terapia Genética , Inhibidores de Crecimiento/genética , Inhibidores de Crecimiento/fisiología , Insulina/sangre , Interleucina-6 , Leptina/sangre , Linfocinas/genética , Linfocinas/fisiología , Tejido Adiposo Pardo/metabolismo , Animales , Glucemia/análisis , Encéfalo/metabolismo , Proteínas Portadoras/genética , Relación Dosis-Respuesta a Droga , Femenino , Inhibidores de Crecimiento/análisis , Canales Iónicos , Factor Inhibidor de Leucemia , Linfocinas/análisis , Proteínas de la Membrana/genética , Proteínas Mitocondriales , Obesidad/sangre , Obesidad/genética , Obesidad/terapia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Proteína Desacopladora 1
10.
Nat Neurosci ; 14(7): 911-8, 2011 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-21642975

RESUMEN

Steroidogenic factor 1 (SF-1)-expressing neurons of the ventromedial hypothalamus (VMH) control energy homeostasis, but the role of insulin action in these cells remains undefined. We show that insulin activates phosphatidylinositol-3-OH kinase (PI3K) signaling in SF-1 neurons and reduces firing frequency in these cells through activation of K(ATP) channels. These effects were abrogated in mice with insulin receptor deficiency restricted to SF-1 neurons (SF-1(ΔIR) mice). Whereas body weight and glucose homeostasis remained the same in SF-1(ΔIR) mice as in controls under a normal chow diet, they were protected from diet-induced leptin resistance, weight gain, adiposity and impaired glucose tolerance. High-fat feeding activated PI3K signaling in SF-1 neurons of control mice, and this response was attenuated in the VMH of SF-1(ΔIR) mice. Mimicking diet-induced overactivation of PI3K signaling by disruption of the phosphatidylinositol-3,4,5-trisphosphate phosphatase PTEN led to increased body weight and hyperphagia under a normal chow diet. Collectively, our experiments reveal that high-fat diet-induced, insulin-dependent PI3K activation in VMH neurons contributes to obesity development.


Asunto(s)
Grasas de la Dieta/efectos adversos , Neuronas/efectos de los fármacos , Obesidad/inducido químicamente , Obesidad/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor de Insulina/metabolismo , Núcleo Hipotalámico Ventromedial/patología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Factores de Edad , Animales , Animales Recién Nacidos , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Calorimetría/métodos , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Proteínas Fluorescentes Verdes/genética , Hipoglucemiantes/farmacología , Técnicas In Vitro , Inyecciones Intraventriculares/métodos , Insulina/farmacología , Leptina/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor Esteroidogénico 1/genética , Factor Esteroidogénico 1/metabolismo , Factores de Tiempo , Tolbutamida/farmacología , Núcleo Hipotalámico Ventromedial/citología
11.
Cell Metab ; 12(1): 88-95, 2010 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-20620998

RESUMEN

Phosphatidyl inositol 3-kinase (PI3K) signaling in the hypothalamus has been implicated in the regulation of energy homeostasis, but the critical brain sites where this intracellular signal integrates various metabolic cues to regulate food intake and energy expenditure are unknown. Here, we show that mice with reduced PI3K activity in the ventromedial hypothalamic nucleus (VMH) are more sensitive to high-fat diet-induced obesity due to reduced energy expenditure. In addition, inhibition of PI3K in the VMH impaired the ability to alter energy expenditure in response to acute high-fat diet feeding and food deprivation. Furthermore, the acute anorexigenic effects induced by exogenous leptin were blunted in the mutant mice. Collectively, our results indicate that PI3K activity in VMH neurons plays a physiologically relevant role in the regulation of energy expenditure.


Asunto(s)
Metabolismo Energético , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Núcleo Hipotalámico Ventromedial/enzimología , Animales , Depresores del Apetito/farmacología , Grasas de la Dieta/farmacología , Homeostasis , Leptina/farmacología , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3
12.
Science ; 297(5582): 843-5, 2002 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-12161655

RESUMEN

Excessive caloric intake is thought to be sensed by the brain, which then activates thermogenesis as a means of preventing obesity. The sympathetic nervous system, through beta-adrenergic receptor (betaAR) action on target tissues, is likely the efferent arm of this homeostatic mechanism. To test this hypothesis, we created mice that lack the three known betaARs (beta-less mice). beta-less mice on a Chow diet had a reduced metabolic rate and were slightly obese. On a high-fat diet, beta-less mice, in contrast to wild-type mice, developed massive obesity that was due entirely to a failure of diet-induced thermogenesis. These findings establish that betaARs are necessary for diet-induced thermogenesis and that this efferent pathway plays a critical role in the body's defense against diet-induced obesity.


Asunto(s)
Dieta , Obesidad/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal , Termogénesis/fisiología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Metabolismo Basal/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Ingestión de Energía , Femenino , Homeostasis/efectos de los fármacos , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Obesidad/sangre , Obesidad/genética , Obesidad/prevención & control , Consumo de Oxígeno/efectos de los fármacos , Fenotipo , Receptores Adrenérgicos beta/genética , Transducción de Señal/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología , Termogénesis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA