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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FEBS J ; 290(4): 1078-1095, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36107440

RESUMEN

Loss of Krev interaction-trapped-1 (KRIT1) expression leads to the development of cerebral cavernous malformations (CCM), a disease in which abnormal blood vessel formation compromises the structure and function of the blood-brain barrier. The role of KRIT1 in regulating endothelial function is well-established. However, several studies have suggested that KRIT1 could also play a role in regulating nonendothelial cell types and, in particular, immune cells. In this study, we generated a mouse model with neutrophil-specific deletion of KRIT1 in order to investigate the effect of KRIT1 deficiency on neutrophil function. Neutrophils isolated from adult Ly6Gtm2621(cre)Arte Krit1flox/flox mice had a reduced ability to attach and spread on the extracellular matrix protein fibronectin and exhibited a subsequent increase in migration. However, adhesion to and migration on ICAM-1 was unchanged. In addition, we used a monomeric, fluorescently-labelled fragment of fibronectin to show that integrin activation is reduced in the absence of KRIT1 expression, though ß1 integrin expression appears unchanged. Finally, neutrophil migration in response to lipopolysaccharide-induced inflammation in the lung was decreased, as shown by reduced cell number and myeloperoxidase activity in lavage samples from Krit1PMNKO mice. Altogether, we show that KRIT1 regulates neutrophil adhesion and migration, likely through regulation of integrin activation, which can lead to altered inflammatory responses in vivo.


Asunto(s)
Adhesión Celular , Movimiento Celular , Proteína KRIT1 , Neutrófilos , Animales , Ratones , Adhesión Celular/genética , Adhesión Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Fibronectinas , Integrina beta1/metabolismo , Proteína KRIT1/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Neutrófilos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo
2.
Methods Mol Biol ; 2152: 259-265, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32524558

RESUMEN

Cerebral cavernous malformation (CCM) is driven by changes in the cerebral microvascular endothelial cell population. Mouse models of CCM have successfully recapitulated the disease in vivo; however, dissection of the disease pathogenesis and molecular mechanism is challenging in vivo due to limited access to the involved tissue in live animals. Therefore, in vitro tissue culture models are required. This protocol is designed to facilitate the isolation of cerebral microvascular endothelial cells from whole murine brain tissue. The protocol utilizes papain for a shorter, single digestion step to maximize cell recovery and viability. Using this technique, we are able to isolate cells from a murine CCM model in which the absence of CCM proteins is driven by Cre-mediated recombination at birth, and results in CCM-like vascular malformations in adult animals.


Asunto(s)
Encéfalo/metabolismo , Separación Celular , Células Endoteliales/metabolismo , Proteína KRIT1/genética , Animales , Barrera Hematoencefálica/metabolismo , Separación Celular/métodos , Modelos Animales de Enfermedad , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Hemangioma Cavernoso del Sistema Nervioso Central/metabolismo , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Mutación
3.
Sci Rep ; 7(1): 8296, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28811547

RESUMEN

The intracellular scaffold KRIT1/CCM1 is an established regulator of vascular barrier function. Loss of KRIT1 leads to decreased microvessel barrier function and to the development of the vascular disorder Cerebral Cavernous Malformation (CCM). However, how loss of KRIT1 causes the subsequent deficit in barrier function remains undefined. Previous studies have shown that loss of KRIT1 increases the production of reactive oxygen species (ROS) and exacerbates vascular permeability triggered by several inflammatory stimuli, but not TNF-α. We now show that endothelial ROS production directly contributes to the loss of barrier function in KRIT1 deficient animals and cells, as targeted antioxidant enzymes reversed the increase in permeability in KRIT1 heterozygous mice as shown by intravital microscopy. Rescue of the redox state restored responsiveness to TNF-α in KRIT1 deficient arterioles, but not venules. In vitro, KRIT1 depletion increased endothelial ROS production via NADPH oxidase signaling, up-regulated Nox4 expression, and promoted NF-κB dependent promoter activity. Recombinant yeast avenanthramide I, an antioxidant and inhibitor of NF-κB signaling, rescued barrier function in KRIT1 deficient cells. However, KRIT1 depletion blunted ROS production in response to TNF-α. Together, our data indicate that ROS signaling is critical for the loss of barrier function following genetic deletion of KRIT1.


Asunto(s)
Endotelio/metabolismo , Proteína KRIT1/deficiencia , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Transducción de Señal , Animales , Antioxidantes/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/genética , Regulación de la Expresión Génica , Proteína KRIT1/genética , Proteína KRIT1/metabolismo , Ratones , Ratones Noqueados , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA