Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Neurobiol ; 59(6): 3755-3766, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35381888

RESUMEN

Circumventricular organs (CVOs), including the mediobasal hypothalamus (MBH), have an incomplete blood-brain barrier (BBB). In this study, we determined if the BBB function in the MBH is modulated by the gut microbiota or by the Toll-like receptor (TLR) adapter proteins TRIF or MyD88 signaling. By injecting mice with Evans blue, a marker for BBB permeability, we show that germ-free (GF) and conventionally raised (CONV-R) mice did not differ in the number of Evans blue-positive cells in MBH. Acute modulation of the gut microbiota did not change the number of Evans blue-positive cells. In contrast, CONV-R Myd88-/- and Trif-/- mice had a reduced number of cells in direct contact to the circulation compared to wildtype (WT) mice. This was accompanied by increased tight junction proteins in the blood vessels in Myd88-/- mice. To further characterize the BBB function, we injected WT and Myd88 -/- CONV-R mice as well as WT GF mice with monosodium glutamate (MSG), a neurotoxin that does not cross the BBB. While MSG caused vast cell death in the MBH in CONV-R and GF WT mice, Myd88 -/- mice were protected from such cell death suggesting that fewer cells are exposed to the neurotoxin in the Myd88 -/- mice. Taken together, our results suggest that MyD88 deficiency, but not gut microbiota depletion, is sufficient to modulate the BBB function in the MBH.


Asunto(s)
Barrera Hematoencefálica , Microbioma Gastrointestinal , Hipotálamo , Factor 88 de Diferenciación Mieloide , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Azul de Evans , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Neurotoxinas/toxicidad , Glutamato de Sodio/toxicidad
2.
Cell Rep ; 35(8): 109163, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34038733

RESUMEN

Mice lacking a microbiota are protected from diet-induced obesity. Previous studies have shown that feeding a Western diet causes hypothalamic inflammation, which in turn can lead to leptin resistance and weight gain. Here, we show that wild-type (WT) mice with depleted gut microbiota, i.e., germ-free (GF) and antibiotic-treated mice, have elevated levels of glucagon-like peptide-1 (GLP-1), are protected against diet-induced hypothalamic inflammation, and have enhanced leptin sensitivity when fed a Western diet. Using GLP-1 receptor (GLP-1R)-deficient mice and pharmacological inhibition of the GLP-1R in WT mice, we demonstrate that intact GLP-1R signaling is required for preventing hypothalamic inflammation and enhancing leptin sensitivity. Furthermore, we show that astrocytes express the GLP-1R, and deletion of the receptor in glial fibrillary acidic protein (GFAP)-expressing cells diminished the antibiotic-induced protection against diet-induced hypothalamic inflammation. Collectively, our results suggest that depletion of the gut microbiota attenuates diet-induced hypothalamic inflammation and enhances leptin sensitivity via GLP-1R-dependent mechanisms.


Asunto(s)
Dieta Occidental/efectos adversos , Microbioma Gastrointestinal/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Hipotálamo/fisiopatología , Inflamación/fisiopatología , Leptina/metabolismo , Obesidad/fisiopatología , Animales , Humanos , Masculino , Ratones
3.
J Innate Immun ; 10(3): 163-171, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29131106

RESUMEN

The gut microbiota has emerged as an environmental factor that modulates the host's energy balance. It increases the host's ability to harvest energy from the digested food, and produces metabolites and microbial products such as short-chain fatty acids, secondary bile acids, and lipopolysaccharides. These metabolites and microbial products act as signaling molecules that modulate appetite, gut motility, energy uptake and storage, and energy expenditure. Several findings suggest that the gut microbiota can affect the development of obesity. Germ-free mice are leaner than conventionally raised mice and they are protected against diet-induced obesity. Furthermore, obese humans and rodents have an altered gut microbiota composition with less phylogeneic diversity compared to lean controls, and transplantation of the gut microbiota from obese subjects to germ-free mice can transfer the obese phenotype. Taken together, these findings indicate a role for the gut microbiota in obesity and suggest that the gut microbiota could be targeted to improve metabolic diseases like obesity. This review focuses on the role of the gut microbiota in energy balance regulation and its potential role in obesity.


Asunto(s)
Metabolismo Energético , Microbioma Gastrointestinal/fisiología , Obesidad/metabolismo , Obesidad/microbiología , Animales , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiología , Leptina/metabolismo , Transducción de Señal , Simbiosis
4.
J Neurosci ; 33(29): 11972-85, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23864684

RESUMEN

Like obesity, prolonged food deprivation induces severe hepatic steatosis; however, the functional significance of this phenomenon is not well understood. In this study, we show that the fall in plasma leptin concentration during fasting is required for the development of hepatic steatosis in mice. Removal of leptin receptors from AGRP neurons diminishes fasting-induced hepatic steatosis. Furthermore, the suppressive effects of leptin on fasting-induced hepatic steatosis are absent in mice lacking the gene encoding agouti-related protein (Agrp), suggesting that this function of leptin is mediated by AGRP. Prolonged fasting leads to suppression of hepatic sympathetic activity, increased expression of acyl CoA:diacylglycerol acyltransferase-2 in the liver, and elevation of hepatic triglyceride content and all of these effects are blunted in the absence of AGRP. AGRP deficiency, despite having no effects on feeding or body adiposity in the free-fed state, impairs triglyceride and ketone body release from the liver during prolonged fasting. Furthermore, reducing CNS Agrp expression in wild-type mice by RNAi protected against the development of hepatic steatosis not only during starvation, but also in response to consumption of a high-fat diet. These findings identify the leptin-AGRP circuit as a critical modulator of hepatic triglyceride stores in starvation and suggest a vital role for this circuit in sustaining the supply of energy from the liver to extrahepatic tissues during periods of prolonged food deprivation.


Asunto(s)
Proteína Relacionada con Agouti/genética , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leptina/metabolismo , Hígado/metabolismo , Receptores de Leptina/genética , Acilcoenzima A/genética , Acilcoenzima A/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Composición Corporal/efectos de los fármacos , Composición Corporal/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Metabolismo Energético/efectos de los fármacos , Hígado Graso/genética , Hígado Graso/metabolismo , Privación de Alimentos/fisiología , Hipotálamo/efectos de los fármacos , Leptina/farmacología , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Norepinefrina/metabolismo , Obesidad/genética , Obesidad/metabolismo , Receptores de Leptina/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(8): E697-706, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23386726

RESUMEN

Chronic consumption of a fat-rich diet leads to attenuation of leptin signaling in hypothalamic neurons, a hallmark feature of cellular leptin resistance. To date, little is known about the temporal and spatial dysregulation of neuronal function under conditions of nutrient excess. We show that agouti-related protein (AgRP)-expressing neurons precede proopiomelanocortin neurons in developing diet-induced cellular leptin resistance. High-fat diet-induced up-regulation of suppressor of cytokine signaling-3 (SOCS3) occurs in AgRP neurons before proopiomelanocortin and other hypothalamic neurons. SOCS3 expression in AgRP neurons increases after 2 d of high-fat feeding, but reduces after switching to a low-fat diet for 1 d. Consistently, transgenic overexpression of SOCS3 in AgRP neurons produces metabolic phenotypes resembling those observed after short-term high-fat feeding. We further show that AgRP neurons are the predominant cell type situated outside the blood-brain barrier in the mediobasal hypothalamus. AgRP neurons are more responsive to low levels of circulating leptin, but they are also more prone to development of leptin resistance in response to a small increase in blood leptin concentrations. Collectively, these results suggest that AgRP neurons are able to sense slight changes in plasma metabolic signals, allowing them to serve as first-line responders to fluctuation of energy intake. Furthermore, modulation of SOCS3 expression in AgRP neurons may play a dynamic and physiological role in metabolic fine tuning in response to short-term changes of nutritional status.


Asunto(s)
Grasas de la Dieta/administración & dosificación , Hipotálamo/fisiopatología , Leptina/fisiología , Neuronas/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Animales , Hipotálamo/citología , Ratones , Neuronas/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Regulación hacia Arriba
6.
Diabetes ; 59(4): 894-906, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20068134

RESUMEN

OBJECTIVE: Hypothalamic leptin resistance is found in most common forms of obesity, such as diet-induced obesity, and is associated with increased expression of suppressor of cytokine signaling 3 (Socs3) in the hypothalamus of diet-induced obese animals. This study aims to determine the functional consequence of Socs3 upregulation on leptin signaling and obesity, and to investigate whether Socs3 upregulation affects energy balance in a cell type-specific way. RESEARCH DESIGN AND METHODS: We generated transgenic mice overexpressing Socs3 in either proopiomelanocortin (POMC) or leptin receptor-expressing neurons, at levels similar to what is observed in diet-induced obesity. RESULTS: Upregulation of Socs3 in POMC neurons leads to impairment of STAT3 and mammalian target of rapamycin (mTOR)-S6K-S6 signaling, with subsequent leptin resistance, obesity, and glucose intolerance. Unexpectedly, Socs3 upregulation in leptin receptor neurons results in increased expression of STAT3 protein in mutant hypothalami, but does not lead to obesity. CONCLUSIONS: Our study establishes that Socs3 upregulation alone in POMC neurons is sufficient to cause leptin resistance and obesity. Socs3 upregulation impairs both STAT3 and mTOR signaling before the onset of obesity. The lack of obesity in mice with upregulated Socs3 in leptin receptor neurons suggests that Socs3's effect on energy balance could be cell type specific. Our study indicates that POMC neurons are important mediators of Socs3's effect on leptin resistance and obesity, but that other cell types or alteration of other signaling regulators could contribute to the development of obesity.


Asunto(s)
Proopiomelanocortina/fisiología , Proteínas Supresoras de la Señalización de Citocinas/genética , Animales , Glucemia/metabolismo , Cartilla de ADN , Femenino , Amplificación de Genes , Genotipo , Hipotálamo/fisiología , Insulina/sangre , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/fisiología , Reacción en Cadena de la Polimerasa , Proopiomelanocortina/genética , Receptores de Leptina/genética , Proteína 3 Supresora de la Señalización de Citocinas , Regulación hacia Arriba
7.
Endocrinology ; 151(2): 671-82, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20022934

RESUMEN

c-Jun-N-terminal kinase (JNK) is a signaling molecule that is activated by proinflammatory signals, endoplasmic reticulum (ER) stress, and other environmental stressors. Although JNK has diverse effects on immunological responses and insulin resistance in peripheral tissues, a functional role for JNK in feeding regulation has not been established. In this study, we show that central inhibition of JNK activity potentiates the stimulatory effects of glucocorticoids on food intake and that this effect is abolished in mice whose agouti-related peptide (AgRP) neurons are degenerated. JNK1-deficient mice feed more upon central administration of glucocorticoids, and glucocorticoid receptor nuclear immunoreactivity is enhanced in the AgRP neurons. JNK inhibition in hypothalamic explants stimulates Agrp expression, and JNK1-deficient mice exhibit increased Agrp expression, heightened hyperphagia, and weight gain during refeeding. Our study shows that JNK1 is a novel regulator of feeding by antagonizing glucocorticoid function in AgRP neurons. Paradoxically, JNK1 mutant mice feed less and lose more weight upon central administration of insulin, suggesting that JNK1 antagonizes insulin function in the brain. Thus, JNK may integrate diverse metabolic signals and differentially regulate feeding under distinct physiological conditions.


Asunto(s)
Apetito/fisiología , Ingestión de Energía/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Proteína Relacionada con Agouti/genética , Animales , Hormona Liberadora de Corticotropina/genética , Ingestión de Energía/efectos de los fármacos , Eliminación de Gen , Homeostasis , Hipotálamo/citología , Hipotálamo/fisiología , Leptina/metabolismo , Leptina/farmacología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Mutantes , Proteína Quinasa 8 Activada por Mitógenos/genética , Neuropéptido Y/genética , Proopiomelanocortina/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Proc Natl Acad Sci U S A ; 106(37): 15932-7, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19805233

RESUMEN

In female mammals including rodents and humans, feeding decreases during the periovulatory period of the ovarian cycle, which coincides with a surge in circulating estrogen levels. Ovariectomy increases food intake, which can be normalized by estrogen treatment at a dose and frequency mimicking those during the estrous cycle. Furthermore, administration of estrogen to rodents potently inhibits food intake. Despite these well-known effects of estrogen, neuronal subtypes that mediate estrogen's anorexigenic effects have not been identified. In this study, we show that changes in hypothalamic expression of agouti-related protein (Agrp) and neuropeptide Y (Npy) coincide with the cyclic changes in feeding across the estrous cycle. These cyclic changes in feeding are abolished in mice with degenerated AgRP neurons even though these mice cycle normally. Central administration of 17beta-estradiol (E2) decreases food intake in controls but not in mice lacking the AgRP neurons. Furthermore, E2 treatment suppresses fasting-induced c-Fos activation in AgRP and NPY neurons and blunts the refeeding response. Surprisingly, although estrogen receptor alpha (ERalpha) is the key mediator of estrogen's anorexigenic effects, we find that expression of ERalpha is completely excluded from AgRP and NPY neurons in the mouse hypothalamus, suggesting that estrogen may regulate these neurons indirectly via presynaptic neurons that express ERalpha. This study indicates that neurons coexpressing AgRP and NPY are functionally required for the cyclic changes in feeding across estrous cycle and that AgRP and NPY neurons are essential mediators of estrogen's anorexigenic function.


Asunto(s)
Proteína Relacionada con Agouti/fisiología , Regulación del Apetito/fisiología , Ciclo Estral/fisiología , Neuropéptido Y/fisiología , Proteína Relacionada con Agouti/deficiencia , Proteína Relacionada con Agouti/genética , Animales , Regulación del Apetito/efectos de los fármacos , Peso Corporal/genética , Peso Corporal/fisiología , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Ciclo Estral/genética , Femenino , Expresión Génica , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuropéptido Y/deficiencia , Neuropéptido Y/genética , Ovariectomía , Ovario/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA