Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(33): eadn5993, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39141732

RESUMEN

Skeletal muscle has gained recognition as an endocrine organ releasing myokines upon contraction during physical exercise. These myokines exert both local and pleiotropic health benefits, underscoring the crucial role of muscle function in countering obesity and contributing to the overall positive effects of exercise on health. Here, we found that exercise activates muscle p38γ, increasing locomotor activity through the secretion of interleukin-15 (IL-15). IL-15 signals in the motor cortex, stimulating locomotor activity. This activation of muscle p38γ, leading to an increase locomotor activity, plays a crucial role in reducing the risk of diabetes and liver steatosis, unveiling a vital muscle-brain communication pathway with profound clinical implications. The correlation between p38γ activation in human muscle during acute exercise and increased blood IL-15 levels highlights the potential therapeutic relevance of this pathway in treating obesity and metabolic diseases. These findings provide valuable insights into the molecular basis of exercise-induced myokine responses promoting physical activity.


Asunto(s)
Ejercicio Físico , Interleucina-15 , Músculo Esquelético , Interleucina-15/metabolismo , Músculo Esquelético/metabolismo , Humanos , Animales , Ejercicio Físico/fisiología , Locomoción , Ratones , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Transducción de Señal , Masculino , Sistema de Señalización de MAP Quinasas , Obesidad/metabolismo
2.
Sci Rep ; 12(1): 5938, 2022 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-35396524

RESUMEN

Vascular smooth muscle cell (VSMC) proliferation is essential for arteriogenesis to restore blood flow after artery occlusion, but the mechanisms underlying this response remain unclear. Based on our previous findings showing increased VSMC proliferation in the neonatal aorta of mice lacking the protease MT4-MMP, we aimed at discovering new players in this process. We demonstrate that MT4-MMP absence boosted VSMC proliferation in vitro in response to PDGF-BB in a cell-autonomous manner through enhanced p38 MAPK activity. Increased phospho-p38 in basal MT4-MMP-null VSMCs augmented the rate of mitochondrial degradation by promoting mitochondrial morphological changes through the co-activator PGC1α as demonstrated in PGC1α-/- VSMCs. We tested the in vivo implications of this pathway in a novel conditional mouse line for selective MT4-MMP deletion in VSMCs and in mice pre-treated with the p38 MAPK activator anisomycin. Priming of p38 MAPK activity in vivo by the absence of the protease MT4-MMP or by anisomycin treatment led to enhanced arteriogenesis and improved flow recovery after femoral artery occlusion. These findings may open new therapeutic opportunities for peripheral vascular diseases.


Asunto(s)
Metaloproteinasa 17 de la Matriz , Proteínas Quinasas p38 Activadas por Mitógenos , Animales , Anisomicina , Proliferación Celular/fisiología , Células Cultivadas , Metaloproteinasa 17 de la Matriz/metabolismo , Ratones , Dinámicas Mitocondriales , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
J Exp Med ; 216(5): 1108-1119, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30944152

RESUMEN

Hepatocellular carcinoma (HCC) is the sixth most common cancer type and the fourth leading cause of cancer-related death. This cancer appears with higher incidence in men and during obesity; however, the specific mechanisms underlying this correlation are unknown. Adipose tissue, a key organ in metabolic syndrome, shows evident gender disparities in the production of adipokines. Levels of the important adipokine adiponectin decrease in men during puberty, as well as in the obese state. Here, we show that this decrease in adiponectin levels is responsible for the increased liver cancer risk in males. We found that testosterone activates the protein JNK in mouse and human adipocytes. JNK-mediated inhibition of adiponectin secretion increases liver cancer cell proliferation, since adiponectin protects against liver cancer development through the activation of AMP-activated protein kinase (AMPK) and p38α. This study provides insight into adipose tissue to liver crosstalk and its gender relation during cancer development, having the potential to guide strategies for new cancer therapeutics.


Asunto(s)
Adiponectina/sangre , Carcinoma Hepatocelular/epidemiología , Cálculos Biliares/sangre , Neoplasias Hepáticas/epidemiología , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/metabolismo , Adiponectina/genética , Tejido Adiposo/metabolismo , Animales , Estudios de Cohortes , Femenino , Cálculos Biliares/cirugía , Humanos , Incidencia , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Obesidad/metabolismo , Factores Sexuales
4.
PLoS Biol ; 16(7): e2004455, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29979672

RESUMEN

Adipose tissue has emerged as an important regulator of whole-body metabolism, and its capacity to dissipate energy in the form of heat has acquired a special relevance in recent years as potential treatment for obesity. In this context, the p38MAPK pathway has arisen as a key player in the thermogenic program because it is required for the activation of brown adipose tissue (BAT) thermogenesis and participates also in the transformation of white adipose tissue (WAT) into BAT-like depot called beige/brite tissue. Here, using mice that are deficient in p38α specifically in adipose tissue (p38αFab-KO), we unexpectedly found that lack of p38α protected against high-fat diet (HFD)-induced obesity. We also showed that p38αFab-KO mice presented higher energy expenditure due to increased BAT thermogenesis. Mechanistically, we found that lack of p38α resulted in the activation of the related protein kinase family member p38δ. Our results showed that p38δ is activated in BAT by cold exposure, and lack of this kinase specifically in adipose tissue (p38δ Fab-KO) resulted in overweight together with reduced energy expenditure and lower body and skin surface temperature in the BAT region. These observations indicate that p38α probably blocks BAT thermogenesis through p38δ inhibition. Consistent with the results obtained in animals, p38α was reduced in visceral and subcutaneous adipose tissue of subjects with obesity and was inversely correlated with body mass index (BMI). Altogether, we have elucidated a mechanism implicated in physiological BAT activation that has potential clinical implications for the treatment of obesity and related diseases such as diabetes.


Asunto(s)
Tejido Adiposo Pardo/enzimología , Tejido Adiposo Pardo/fisiología , Proteína Quinasa 13 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Termogénesis , Adipocitos Marrones/enzimología , Adulto , Animales , Índice de Masa Corporal , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/prevención & control , Dieta , Metabolismo Energético , Activación Enzimática , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 13 Activada por Mitógenos/metabolismo , Modelos Biológicos , Obesidad/enzimología , Obesidad/prevención & control , Proteína Desacopladora 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA