Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Endocrinology ; 164(7)2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37279930

RESUMEN

When mammals are exposed to a warm environment, overheating is prevented by activation of "warm-responsive" neurons (WRNs) in the hypothalamic preoptic area (POA) that reduce thermogenesis while promoting heat dissipation. Heat exposure also impairs glucose tolerance, but whether this also results from activation of POA WRNs is unknown. To address this question, we sought in the current work to determine if glucose intolerance induced by heat exposure can be attributed to activation of a specific subset of WRNs that express pituitary adenylate cyclase-activating peptide (ie, POAPacap neurons). We report that when mice are exposed to an ambient temperature sufficiently warm to activate POAPacap neurons, the expected reduction of energy expenditure is associated with glucose intolerance, and that these responses are recapitulated by chemogenetic POAPacap neuron activation. Because heat-induced glucose intolerance was not blocked by chemogenetic inhibition of POAPacap neurons, we conclude that POAPacap neuron activation is sufficient, but not required, to explain the impairment of glucose tolerance elicited by heat exposure.


Asunto(s)
Hipotálamo , Área Preóptica , Ratones , Masculino , Animales , Área Preóptica/fisiología , Homeostasis , Hipotálamo/fisiología , Regulación de la Temperatura Corporal/fisiología , Neuronas/fisiología , Glucosa , Mamíferos
2.
PLoS One ; 12(2): e0172912, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28245284

RESUMEN

BACKGROUND: Widely used as a weight loss supplement, trans-10,cis-12 conjugated linoleic acid (10,12 CLA) promotes fat loss in obese mice and humans, but has also been associated with insulin resistance. OBJECTIVE: We therefore sought to directly compare weight loss by 10,12 CLA versus caloric restriction (CR, 15-25%), an acceptable healthy method of weight loss, to determine how 10,12 CLA-mediated weight loss fails to improve glucose metabolism. METHODS: Obese mice with characteristics of human metabolic syndrome were either supplemented with 10,12 CLA or subjected to CR to promote weight loss. Metabolic endpoints such as energy expenditure, glucose and insulin tolerance testing, and trunk fat distribution were measured. RESULTS: By design, 10,12 CLA and CR caused equivalent weight loss, with greater fat loss by 10,12 CLA accompanied by increased energy expenditure, reduced respiratory quotient, increased fat oxidation, accumulation of alternatively activated macrophages, and browning of subcutaneous white adipose tissue (WAT). Moreover, 10,12 CLA-supplemented mice better defended their body temperature against a cold challenge. However, 10,12 CLA concurrently induced the detrimental loss of subcutaneous WAT without reducing visceral WAT, promoted reduced plasma and WAT adipokine levels, worsened hepatic steatosis, and failed to improve glucose metabolism. Obese mice undergoing CR were protected from subcutaneous-specific fat loss, had improved hepatic steatosis, and subsequently showed the expected improvements in WAT adipokines, glucose metabolism and WAT inflammation. CONCLUSIONS: These results suggest that 10,12 CLA mediates the preferential loss of subcutaneous fat that likely contributes to hepatic steatosis and maintained insulin resistance, despite significant weight loss and WAT browning in mice. Collectively, we have shown that weight loss due to 10,12 CLA supplementation or CR results in dramatically different metabolic phenotypes, with the latter promoting a healthier form of weight loss.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Restricción Calórica , Glucosa/metabolismo , Ácidos Linoleicos Conjugados/farmacología , Grasa Subcutánea/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Femenino , Homeostasis , Masculino , Ratones , Ratones Obesos , Grasa Subcutánea/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos , Pérdida de Peso/fisiología
3.
Pediatr Res ; 69(3): 230-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21372758

RESUMEN

Patients with craniopharyngioma (CP), a tumor located in the pituitary and/or hypothalamus, are susceptible to developing obesity and many metabolic complications. The study aim was to create a rodent model that mimics the complex neuroanatomical and metabolic disturbances commonly seen in obese CP patients. We compared the metabolic phenotype of animals with three distinct types of hypothalamic lesions: 1) destruction of the arcuate nucleus (ARC) induced by monosodium glutamate (MSG), 2) electrolytic lesion of the adjacent ventromedial nucleus (VMN) alone, 3) both the VMN and dorsomedial nucleus (DMN), or a 4) combined medial hypothalamic lesion (CMHL) affecting the VMN, DMN, and the ARC. Only the CMHL model exhibited all key features observed in patients with hypothalamic obesity induced by CP. These features included excessive weight gain due to increased adiposity, increased food intake, and pronounced hyperinsulinemia and hyperleptinemia. Similar to characteristics of patients with CP, CMHL animals exhibited reduced plasma levels of alpha-melanocyte stimulating hormone and reduced ambulatory activity compared with weight-matched controls. Therefore, the CMHL model best mimics the complex metabolic abnormalities observed in obese CP patients compared with lesions to other hypothalamic areas and provides a foundation for future pharmacological approaches to treat obesity in children with hypothalamic damage.


Asunto(s)
Craneofaringioma/complicaciones , Modelos Animales de Enfermedad , Neoplasias Hipotalámicas/complicaciones , Obesidad/etiología , Neoplasias Hipofisarias/complicaciones , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/patología , Peso Corporal , Niño , Craneofaringioma/patología , Núcleo Hipotalámico Dorsomedial/metabolismo , Núcleo Hipotalámico Dorsomedial/patología , Ingestión de Alimentos , Metabolismo Energético , Femenino , Homeostasis , Humanos , Neoplasias Hipotalámicas/patología , Hipotálamo/anatomía & histología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Neoplasias Hipofisarias/patología , Embarazo , Ratas , Ratas Sprague-Dawley , Glutamato de Sodio/efectos adversos , Núcleo Hipotalámico Ventromedial/metabolismo , Núcleo Hipotalámico Ventromedial/patología
4.
Am J Physiol Endocrinol Metab ; 300(2): E392-401, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21062956

RESUMEN

Mechanisms regulating spontaneous physical activity remain poorly characterized despite evidence of influential genetic and acquired factors. We evaluated ambulatory activity and wheel running in leptin-deficient ob/ob mice and in wild-type mice rendered hypoleptinemic by fasting in both the presence and absence of subcutaneous leptin administration. In ob/ob mice, leptin treatment to plasma levels characteristic of wild-type mice acutely increased both ambulatory activity (by 4,000 ± 200 beam breaks/dark cycle, P < 0.05) and total energy expenditure (TEE; by 0.11 ± 0.01 kcal/h during the dark cycle, P < 0.05) in a dose-dependent manner and acutely increased wheel running (+350%, P < 0.05). Fasting potently increased ambulatory activity and wheel running in wild-type mice (AA: +25%, P < 0.05; wheel running: +80%, P < 0.05), and the effect of fasting was more pronounced in ob/ob mice (AA: +400%, P < 0.05; wheel running: +1,600%, P < 0.05). However, unlike what occurred in ad libitum-fed ob/ob mice, physiological leptin replacement attenuated or prevented fasting-induced increases of ambulatory activity and wheel running in both wild-type and ob/ob mice. Thus, plasma leptin is a physiological regulator of spontaneous physical activity, but the nature of leptin's effect on activity is dependent on food availability.


Asunto(s)
Leptina/fisiología , Actividad Motora/fisiología , Carrera/fisiología , Animales , Composición Corporal , Calorimetría Indirecta , Oscuridad , Relación Dosis-Respuesta a Droga , Metabolismo Energético , Ensayo de Inmunoadsorción Enzimática , Ayuno/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Leptina/farmacología , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Actividad Motora/efectos de los fármacos , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Consumo de Oxígeno/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Am J Physiol Endocrinol Metab ; 299(1): E47-53, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20371733

RESUMEN

In peripheral tissues, the link between obesity and insulin resistance involves low-grade inflammation induced by macrophage activation and proinflammatory cytokine signaling. Since proinflammatory cytokines are also induced in the hypothalamus of animals placed on a high-fat (HF) diet and can inhibit neuronal signal transduction pathways required for normal energy homeostasis, hypothalamic inflammation is hypothesized to contribute to the pathogenesis of diet-induced obesity (DIO). We addressed this hypothesis by perturbing the inflammatory milieu of the hypothalamus in adult male Wistar rats using intracerebroventricular (icv) administration of interleukin-4 (IL-4), a Th2 cytokine that promotes alternative activation (M2) of macrophages and microglia. During HF feeding, icv IL-4 administration increased hypothalamic proinflammatory cytokine gene expression and caused excess weight gain. Intracerebroventricular pretreatment with PS1145, an inhibitor of IKKbeta (a key intracellular mediator of inflammatory signaling), blocked both IL-4 effects, suggesting a causal relationship between IL-4-induced weight gain and hypothalamic inflammation. These observations add to growing evidence linking hypothalamic inflammation to obesity pathogenesis.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Metabolismo Energético/fisiología , Hipotálamo/fisiopatología , Inflamación/fisiopatología , Interleucina-4/farmacología , Obesidad/fisiopatología , Animales , Glucemia/análisis , Peso Corporal/fisiología , Grasas de la Dieta/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , Insulina/sangre , Leptina/sangre , Activación de Macrófagos/fisiología , Masculino , Piridinas/farmacología , Ratas , Ratas Wistar , Organismos Libres de Patógenos Específicos
6.
Endocrinology ; 150(12): 5362-72, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19819945

RESUMEN

By activating the Toll-like receptor 4-nuclear factor-kappaB signal transduction pathway, the bacterial endotoxin lipopolysaccharide (LPS) induces anorexia, weight loss, fever, and other components of the sickness response. By comparison, the hormones leptin and insulin cause anorexia without sickness via a central mechanism involving the phosphatidylinositol-3 kinase signaling pathway. In the current study, we investigated whether a common Toll-like receptor 4 and phosphatidylinositol-3 kinase signaling intermediate, atypical protein kinase Czeta/lambda (aPKC), contributes to changes of energy balance induced by these stimuli. Immunohistochemistry analysis revealed that aPKC is expressed in the arcuate and paraventricular nuclei of the hypothalamus, key sites of leptin, insulin, and LPS action. Although administration of LPS, insulin, and leptin each acutely increased hypothalamic aPKC activity at doses that also reduce food intake, LPS treatment caused over 10-fold greater activation of hypothalamic a PKC signaling than that induced by leptin or insulin. Intracerebroventricular pretreatment with an aPKC inhibitor blocked anorexia induced by LPS but not insulin or leptin. Similarly, LPS-induced hypothalamic inflammation (as judged by induction of proinflammatory cytokine gene expression) and neuronal activation in the paraventricular nucleus (as judged by c-fos induction) were reduced by central aPKC inhibition. Although intracerebroventricular aPKC inhibitor administration also abolished LPS-induced fever, it had no effect on sickness-related hypoactivity or weight loss. We conclude that although hypothalamic aPKC signaling is not required for food intake inhibition by insulin or leptin, it plays a key role in inflammatory anorexia and fever induced by LPS.


Asunto(s)
Anorexia/enzimología , Hipotálamo/efectos de los fármacos , Lipopolisacáridos/toxicidad , Proteína Quinasa C/metabolismo , Animales , Anorexia/inducido químicamente , Línea Celular , Citocinas/genética , Ingestión de Alimentos/efectos de los fármacos , Fiebre/inducido químicamente , Fiebre/enzimología , Expresión Génica/efectos de los fármacos , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Inmunohistoquímica , Inflamación/enzimología , Inflamación/patología , Insulina/farmacología , Isoenzimas/metabolismo , Leptina/farmacología , Masculino , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Endocrinology ; 149(12): 6053-64, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18687778

RESUMEN

Energy homeostasis involves central nervous system integration of afferent inputs that coordinately regulate food intake and energy expenditure. Here, we report that adult homozygous TNFalpha converting enzyme (TACE)-deficient mice exhibit one of the most dramatic examples of hypermetabolism yet reported in a rodent system. Because this effect is not matched by increased food intake, mice lacking TACE exhibit a lean phenotype. In the hypothalamus of these mice, neurons in the arcuate nucleus exhibit intact responses to reduced fat mass and low circulating leptin levels, suggesting that defects in other components of the energy homeostasis system explain the phenotype of Tace(DeltaZn/DeltaZn) mice. Elevated levels of uncoupling protein-1 in brown adipose tissue from Tace(DeltaZn/DeltaZn) mice when compared with weight-matched controls suggest that deficient TACE activity is linked to increased sympathetic outflow. These findings collectively identify a novel and potentially important role for TACE in energy homeostasis.


Asunto(s)
Proteínas ADAM/deficiencia , Metabolismo Energético/fisiología , Delgadez/fisiopatología , Proteínas ADAM/genética , Proteína ADAM17 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Western Blotting , Diferenciación Celular/fisiología , Ingestión de Alimentos/fisiología , Grasas/metabolismo , Hipotálamo/metabolismo , Inmunohistoquímica , Canales Iónicos/metabolismo , Leptina/sangre , Masculino , Ratones , Ratones Mutantes , Proteínas Mitocondriales/metabolismo , Actividad Motora/fisiología , Fenotipo , Delgadez/sangre , Delgadez/genética , Proteína Desacopladora 1
8.
Endocrinology ; 148(11): 5230-7, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17673516

RESUMEN

Systemic inflammatory stimuli cause anorexia and weight loss by disrupting the physiological regulation of energy balance. Mice lacking MyD88, an intracellular mediator of signal transduction activated by Toll-like receptor 4 or IL-1beta receptors, are resistant to anorexia induced by the bacterial endotoxin lipopolysaccharide (LPS), despite a significant circulating cytokine response. Thus, we hypothesized that induction of a peripheral inflammatory response is insufficient to cause LPS-induced anorexia when MyD88 signaling in the central nervous system and other tissues is absent. To test this hypothesis, we used bone marrow transplantation (BMT) to determine if LPS-induced anorexia can be restored to MyD88-deficient mice by reconstituting their bone marrow with wild-type (WT) immune cells. We found that restoring WT circulating immune cells to mice lacking MyD88 conferred only a mild, short-lived anorexia in response to LPS, such that food intake was fully normalized by 20 h post injection (LPS 4.1 +/- 0.5 g vs. vehicle 4.3 +/- 0.3 g), whereas LPS-induced anorexia was profound and sustained in WT controls after either autologous BMT or sham BMT. Similarly, LPS-mediated induction of hypothalamic mRNA encoding IL-1beta and TNFalpha was robust in both WT control groups but was absent in chimeric MyD88 mice, despite comparable peripheral inflammatory responses across the three groups. We conclude that LPS reduces food intake via a mechanism dependent on MyD88 signaling within brain and/or other tissues and that in the absence of this effect, robust stimulation of circulating immune cells cannot induce sustained anorexia.


Asunto(s)
Anorexia/inducido químicamente , Hipotálamo/fisiología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/farmacología , Transducción de Señal/fisiología , Bazo/fisiología , Animales , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Trasplante de Médula Ósea/fisiología , Células Cultivadas , Ingestión de Alimentos/efectos de los fármacos , Femenino , Hipotálamo/metabolismo , Sistema Inmunológico/metabolismo , Sistema Inmunológico/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Bazo/metabolismo
9.
Endocrinology ; 147(9): 4445-53, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16777969

RESUMEN

Systemic inflammatory signals can disrupt the physiological regulation of energy balance, causing anorexia and weight loss. In the current studies, we investigated whether MyD88, the primary, but not exclusive, intracellular signal transduction pathway for Toll-like receptor 4 and IL-1 receptor I, is necessary for anorexia and weight loss to occur in response to stimuli that activate these key innate immune receptors. Our findings demonstrate that the absence of MyD88 signaling confers complete protection against anorexia induced by either lipopolysaccharide (LPS) (20 h food intake in MyD88-/- mice 5.4 +/- 0.3 vs. 3.3 +/- 0.4 g in MyD88+/+ control mice, P < 0.001) or IL-1 beta (20 h food intake in MyD88-/- mice 4.9 +/- 0.5 vs. 4.0 +/- 0.3 g in MyD88+/+ control mice, P < 0.001). However, absent MyD88 signaling does not prevent these inflammatory mediators from causing weight loss (LPS, -0.4 +/- 0.1 g; IL1 beta, -0.1 +/- 0.1 g, both P < 0.01 vs. vehicle-injected MyD88-/- mice, +0.4 +/- 0.2 g). Furthermore, LPS-induced weight loss occurs in the absence of adipsia, fever, or hypothalamus-pituitary-adrenal axis activation in MyD88-deficient mice. In addition, the peripheral inflammatory response to LPS is surprisingly intact in mice lacking MyD88. Together, these observations indicate that LPS reduces food intake via a mechanism that is dissociated from its effect on peripheral cytokine production, and whereas the presence of circulating proinflammatory cytokines per se is insufficient to cause anorexia in the absence of MyD88 signaling, it may contribute to LPS-induced weight loss.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Anorexia/fisiopatología , Interleucina-1/farmacología , Lipopolisacáridos/farmacología , Pérdida de Peso/fisiología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Animales , Anorexia/etiología , Anorexia/prevención & control , Temperatura Corporal/efectos de los fármacos , Corticosterona/sangre , Citocinas/sangre , Citocinas/genética , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Metabolismo Energético/efectos de los fármacos , Femenino , Hipotálamo/química , Interleucina-6/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide , Neuropéptidos/genética , ARN Mensajero/análisis , Receptores de Interleucina-1/fisiología , Transducción de Señal , Bazo/química , Receptor Toll-Like 4/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Pérdida de Peso/efectos de los fármacos
10.
Peptides ; 27(2): 265-73, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16274848

RESUMEN

In the current study we sought to determine whether hypothalamic IL-1beta is regulated by melanocortin signaling and if melanocortin-induced changes in energy balance are dependent on IL-1beta. A melanocortin agonist, MTII, increased hypothalamic IL-1beta mRNA levels by two-fold, whereas a melanocortin antagonist, SHU9119, blunted lipopolysaccharide (LPS)-mediated increase of hypothalamic IL-1beta content. Pharmacological or genetic disruption of IL-1 receptor signaling prevented MTII-mediated reductions in locomotor activity, but did not reduce MTII-induced anorexia. These data suggest a potential role for central melanocortins in mediating the decrease of ambulation characteristic of the 'sickness' response.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Interleucina-1/fisiología , Hormonas Estimuladoras de los Melanocitos/fisiología , Animales , Homeostasis , Hipotálamo/química , Interleucina-1/genética , Masculino , Ratones , Ratones Noqueados , Movimiento , Ratas , Ratas Wistar , Transducción de Señal
11.
PLoS Biol ; 3(12): e415, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16296893

RESUMEN

Normal aging in humans and rodents is accompanied by a progressive increase in adiposity. To investigate the role of hypothalamic neuronal circuits in this process, we used a Cre-lox strategy to create mice with specific and progressive degeneration of hypothalamic neurons that express agouti-related protein (Agrp) or proopiomelanocortin (Pomc), neuropeptides that promote positive or negative energy balance, respectively, through their opposing effects on melanocortin receptor signaling. In previous studies, Pomc mutant mice became obese, but Agrp mutant mice were surprisingly normal, suggesting potential compensation by neuronal circuits or genetic redundancy. Here we find that Pomc-ablation mice develop obesity similar to that described for Pomc knockout mice, but also exhibit defects in compensatory hyperphagia similar to what occurs during normal aging. Agrp-ablation female mice exhibit reduced adiposity with normal compensatory hyperphagia, while animals ablated for both Pomc and Agrp neurons exhibit an additive interaction phenotype. These findings provide new insight into the roles of hypothalamic neurons in energy balance regulation, and provide a model for understanding defects in human energy balance associated with neurodegeneration and aging.


Asunto(s)
Metabolismo Energético , Hipotálamo/patología , Hipotálamo/fisiopatología , Degeneración Nerviosa/fisiopatología , Proteína Relacionada con Agouti , Animales , Peso Corporal/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ingestión de Alimentos , Eliminación de Gen , Genes Reporteros/genética , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Proopiomelanocortina/deficiencia , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , ARN Mensajero/genética
12.
Am J Physiol Endocrinol Metab ; 287(6): E1107-13, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15304373

RESUMEN

Interleukin-1beta (IL-1beta) is synthesized in a variety of tissues, including the hypothalamus, where it is implicated in the control of food intake. The current studies were undertaken to investigate whether hypothalamic IL-1beta gene expression is subject to physiological regulation by leptin and glucocorticoids (GCs), key hormones involved in energy homeostasis. Adrenalectomy (ADX) increased hypothalamic IL-1beta mRNA levels twofold, measured by real-time PCR (P < 0.05 vs. sham-operated controls), and this effect was blocked by subcutaneous infusion of a physiological dose of corticosterone. Conversely, hypothalamic IL-1beta mRNA levels were reduced by 30% in fa/fa (Zucker) rats, a model of genetic obesity caused by leptin receptor mutation (P = 0.01 vs. lean littermates), and the effect of ADX to increase hypothalamic IL-1beta mRNA levels in fa/fa rats (P = 0.02) is similar to that seen in normal animals. Moreover, fasting for 48 h (which lowers leptin and raises corticosterone levels) reduced hypothalamic IL-1beta mRNA levels by 30% (P = 0.02), and this decrease was fully reversed by refeeding for 12 h. Thus leptin and GCs exert opposing effects on hypothalamic IL-1beta gene expression, and corticosterone plays a physiological role to limit expression of this cytokine in both the presence and absence of intact leptin signaling. Consistent with this hypothesis, systemic leptin administration to normal rats (2 mg/kg ip) increased hypothalamic IL-1beta mRNA levels twofold (P < 0.05 vs. vehicle), an effect similar to that of ADX. These data support a model in which expression of hypothalamic IL-1beta is subject to opposing physiological regulation by corticosterone and leptin.


Asunto(s)
Corticosterona/fisiología , Hipotálamo/metabolismo , Interleucina-1/metabolismo , Leptina/fisiología , Adrenalectomía , Alimentación Animal , Animales , Peso Corporal , Corticosterona/sangre , Corticosterona/farmacología , Ingestión de Alimentos , Metabolismo Energético/fisiología , Ayuno , Expresión Génica/fisiología , Interleucina-1/genética , Leptina/sangre , Leptina/farmacología , Masculino , Obesidad/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Ratas Zucker
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA