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

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
FASEB J ; : fj201800109R, 2018 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-29763381

RESUMEN

Asthma affects ∼300 million people worldwide. Despite multiple treatment options, asthma treatment remains unsatisfactory in a subset of patients. Airway obstruction is a hallmark of allergic asthma and is largely due to airway smooth muscle hypercontractility induced by airway inflammation. Identification of molecular pathways that regulate airway smooth muscle hypercontractility is of considerable therapeutic interest. We previously identified roles for milk fat globule epidermal growth factor-like 8 (Mfge8) in opposing the effects of allergic inflammation on increasing airway smooth muscle contractile force. In this study, we delineate the signaling pathway by which Mfge8 mediates these effects. By using genetic and pharmacologic approaches, we show that the α8ß1 integrin and the phosphatase and tensin homolog (PTEN) mediate the effects of Mfge8 on preventing IL-13-induced increases in airway contractility. Tracheal rings from mice with smooth muscle-specific deletion of α8ß1 or PTEN have enhanced contraction in response to treatment with IL-13. Enhanced IL-13-induced tracheal ring contraction in Mfge8-/- mice was abolished by treatment with the PI3K inhibitor. Mechanistically, IL-13 induces ubiquitination and degradation of PTEN protein. Our findings identify a role for the Mfge8-α8ß1-PTEN pathway in regulating the force of airway smooth muscle contraction in the setting of allergic inflammation.-Khalifeh-Soltani, A., Gupta, D., Ha, A., Podolsky, M. J., Datta, R., Atabai, K. The Mfge8-α8ß1-PTEN pathway regulates airway smooth muscle contraction in allergic inflammation.

2.
Am J Physiol Lung Cell Mol Physiol ; 315(3): L360-L370, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29792348

RESUMEN

Although extensive work has delineated many of the mechanisms of extracellular matrix (ECM) production, far less is known about pathways that regulate ECM degradation. This is particularly true of cellular internalization and degradation of matrix, which play an underappreciated role in ECM metabolism and lung fibrosis. For example, genetic perturbation of this pathway leads to exacerbated fibrosis in experimental animal models. In this work, we present the results of an unbiased screen of Drosophila phagocytes that yielded multiple genes that, when silenced, led to increased collagen uptake. We further describe the function of cell division cycle 7 kinase (CDC7) as a specific suppressor of collagen uptake. We show that the genetic or pharmacological inhibition of CDC7 results in increased expression of the collagen endocytic receptor Endo180. Chromobox 5 (CBX5) is a putative target of CDC7, and genetic silencing of CBX5 also results in increased Endo180 and collagen uptake. Finally, CRISPR-mediated activation of Endo180 expression results in increased collagen uptake, suggesting that CDC7 regulates collagen internalization through increased Endo180 expression. Targeting the regulatory elements of the collagen degradative machinery may be a useful therapeutic approach in diseases of fibrosis or malignancy.


Asunto(s)
Colágeno/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteolisis , Animales , Línea Celular , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Colágeno/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibrosis , Regulación Enzimológica de la Expresión Génica , Proteínas Serina-Treonina Quinasas/genética , Receptores Mitogénicos/biosíntesis , Receptores Mitogénicos/genética
3.
JCI Insight ; 1(18): e87418, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27812539

RESUMEN

The small intestine has an underappreciated role as a lipid storage organ. Under conditions of high dietary fat intake, enterocytes can minimize the extent of postprandial lipemia by storing newly absorbed dietary fat in cytoplasmic lipid droplets. Lipid droplets can be subsequently mobilized for the production of chylomicrons. The mechanisms that regulate this process are poorly understood. We report here that the milk protein Mfge8 regulates hydrolysis of cytoplasmic lipid droplets in enterocytes after interacting with the αvß3 and αvß5 integrins. Mice deficient in Mfge8 or the αvß3 and αvß5 integrins accumulate excess cytoplasmic lipid droplets after a fat challenge. Mechanistically, interruption of the Mfge8-integrin axis leads to impaired enterocyte intracellular triglyceride hydrolase activity in vitro and in vivo. Furthermore, Mfge8 increases triglyceride hydrolase activity through a PI3 kinase/mTORC2-dependent signaling pathway. These data identify a key role for Mfge8 and the αvß3 and αvß5 integrins in regulating enterocyte lipid processing.


Asunto(s)
Antígenos de Superficie/metabolismo , Enterocitos/enzimología , Hidrolasas/metabolismo , Metabolismo de los Lípidos , Proteínas de la Leche/metabolismo , Animales , Células CACO-2 , Quilomicrones , Humanos , Integrina alfaVbeta3/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Vitronectina/metabolismo , Triglicéridos
4.
Elife ; 52016 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-27092791

RESUMEN

Coordinated gastrointestinal smooth muscle contraction is critical for proper nutrient absorption and is altered in a number of medical disorders. In this work, we demonstrate a critical role for the RGD-binding integrin α8ß1 in promoting nutrient absorption through regulation of gastrointestinal motility. Smooth muscle-specific deletion and antibody blockade of α8 in mice result in enhanced gastric antral smooth muscle contraction, more rapid gastric emptying, and more rapid transit of food through the small intestine leading to malabsorption of dietary fats and carbohydrates as well as protection from weight gain in a diet-induced model of obesity. Mechanistically, ligation of α8ß1 by the milk protein Mfge8 reduces antral smooth muscle contractile force by preventing RhoA activation through a PTEN-dependent mechanism. Collectively, our results identify a role for α8ß1 in regulating gastrointestinal motility and identify α8 as a potential target for disorders characterized by hypo- or hyper-motility.


Asunto(s)
Adsorción , Antígenos de Superficie/metabolismo , Alimentos , Integrinas/metabolismo , Proteínas de la Leche/metabolismo , Fosfohidrolasa PTEN/metabolismo , Animales , Motilidad Gastrointestinal , Tracto Gastrointestinal/fisiología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA