Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS One ; 13(1): e0190205, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29293568

RESUMEN

Alzheimer's disease is a neurodegenerative disorder that affects the central nervous system. In this study, we characterized and examined the early metabolic changes in the triple transgenic mouse AD model (3xtg-AD), and their relationship with the hypothalamus, a key regulator of metabolism in the central nervous system. We observed that the 3xtg-AD model exhibited significantly higher oxygen consumption as well as food intake before reported amyloid plaque formation, indicating that metabolic abnormalities occurred at early onset in the 3xtg-AD model compared with their counterparts. Analysis of gene expression in the hypothalamus indicated increased mRNA expression of inflammation- and apoptosis-related genes, as well as decreased gene expression of Agouti-related protein (AgRP) and Melanocortin 4 receptor (MC4R) at 12 weeks of age. Immunofluorescence analysis revealed that pro-opiomelanocortin (POMC) and NPY-expressing neurons decreased at 24 weeks in the 3xtg-AD model. Four weeks of voluntary exercise were sufficient to reverse the gene expression of inflammation and apoptotic markers in the hypothalamus, six weeks of exercise improved glucose metabolism, moreover, 8 weeks of voluntary exercise training attenuated apoptosis and augmented POMC and NPY-expressing neuronal populations in the hypothalamus compared to the control group. Our results indicated that early onset of metabolic abnormalities may contribute to the pathology of AD, which is associated with increased inflammation as well as decreased neuronal population and key neuropeptides in the hypothalamus. Furthermore, early intervention by voluntary exercise normalized hypothalamic inflammation and neurodegeneration as well as glucose metabolism in the 3xtg-AD model. The data, taken as a whole, suggests a hypothalamic-mediated mechanism where exercise prevents the progression of dementia and of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Modelos Animales de Enfermedad , Hipotálamo/patología , Condicionamiento Físico Animal , Animales , Biomarcadores/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Hipotálamo/metabolismo , Etiquetado Corte-Fin in Situ , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Proopiomelanocortina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
J Endocrinol ; 229(2): 109-22, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26931136

RESUMEN

Exercise plays a critical role in regulating glucose homeostasis and body weight. However, the mechanism of exercise on metabolic functions associated with the CNS has not been fully understood. C57BL6 male mice (n=45) were divided into three groups: normal chow diet, high-fat diet (HFD) treatment, and HFD along with voluntary running wheel exercise training for 12 weeks. Metabolic function was examined by the Comprehensive Lab Animal Monitoring System and magnetic resonance imaging; phenotypic analysis included measurements of body weight, food intake, glucose and insulin tolerance tests, as well as insulin and leptin sensitivity studies. By immunohistochemistry, the amount changes in the phosphorylation of signal transducer and activator of transcription 3, neuronal proliferative maker Ki67, apoptosis positive cells as well as pro-opiomelanocortin (POMC)-expressing neurons in the arcuate area of the hypothalamus was identified. We found that 12 weeks of voluntary exercise training partially reduced body weight gain and adiposity induced by an HFD. Insulin and leptin sensitivity were enhanced in the exercise training group verses the HFD group. Furthermore, the HFD-impaired POMC-expressing neuron is remarkably restored in the exercise training group. The restoration of POMC neuron number may be due to neuroprotective effects of exercise on POMC neurons, as evidenced by altered proliferation and apoptosis. In conclusion, our data suggest that voluntary exercise training improves metabolic symptoms induced by HFD, in part through protected POMC-expressing neuron from HFD and enhanced leptin signaling in the hypothalamus that regulates whole-body energy homeostasis.


Asunto(s)
Hipotálamo/fisiopatología , Obesidad/fisiopatología , Condicionamiento Físico Animal/fisiología , Adiposidad , Animales , Proliferación Celular , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Hipotálamo/patología , Resistencia a la Insulina , Leptina/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Neuronas/metabolismo , Neuronas/patología , Obesidad/patología , Obesidad/terapia , Esfuerzo Físico/fisiología , Proopiomelanocortina/metabolismo , Transducción de Señal , Aumento de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA