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

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Immunity ; 40(1): 66-77, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24412613

RESUMEN

Acute intestinal inflammation involves early accumulation of neutrophils (PMNs) followed by either resolution or progression to chronic inflammation. Based on recent evidence that mucosal metabolism influences disease outcomes, we hypothesized that transmigrating PMNs influence the transcriptional profile of the surrounding mucosa. Microarray studies revealed a cohort of hypoxia-responsive genes regulated by PMN-epithelial crosstalk. Transmigrating PMNs rapidly depleted microenvironmental O2 sufficiently to stabilize intestinal epithelial cell hypoxia-inducible factor (HIF). By utilizing HIF reporter mice in an acute colitis model, we investigated the relative contribution of PMNs and the respiratory burst to "inflammatory hypoxia" in vivo. CGD mice, lacking a respiratory burst, developed accentuated colitis compared to control, with exaggerated PMN infiltration and diminished inflammatory hypoxia. Finally, pharmacological HIF stabilization within the mucosa protected CGD mice from severe colitis. In conclusion, transcriptional imprinting by infiltrating neutrophils modulates the host response to inflammation, via localized O2 depletion, resulting in microenvironmental hypoxia and effective inflammatory resolution.


Asunto(s)
Colitis/inmunología , Hipoxia/inmunología , Membrana Mucosa/metabolismo , Neutrófilos/patología , Animales , Comunicación Celular , Movimiento Celular , Células Cultivadas , Microambiente Celular , Colitis/inducido químicamente , Colon/patología , Modelos Animales de Enfermedad , Hipoxia/inducido químicamente , Factor 1 Inducible por Hipoxia/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Membrana Mucosa/patología , NADPH Oxidasa 2 , NADPH Oxidasas/genética , Estrés Oxidativo , Oxígeno/metabolismo , Estabilidad Proteica/efectos de los fármacos , Migración Transendotelial y Transepitelial
2.
Proc Natl Acad Sci U S A ; 107(32): 14298-303, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20660763

RESUMEN

Resolvin-E1 (RvE1) has been demonstrated to promote inflammatory resolution in numerous disease models. Given the importance of epithelial cells to coordination of mucosal inflammation, we hypothesized that RvE1 elicits an epithelial resolution signature. Initial studies revealed that the RvE1-receptor (ChemR23) is expressed on intestinal epithelial cells (IECs) and that microarray profiling of cells exposed to RvE1 revealed regulation of inflammatory response gene expression. Notably, RvE1 induced intestinal alkaline phosphatase (ALPI) expression and significantly enhanced epithelial ALPI enzyme activity. One role recently attributed to ALPI is the detoxification of bacterial LPS. In our studies, RvE1-exposed epithelia detoxified LPS (assessed by attenuation of NF-kappaB signaling). Furthermore, in epithelial-bacterial interaction assays, we determined that ALPI retarded the growth of Escherichia coli. To define these features in vivo, we used a murine dextran sulfate sodium (DSS) model of colitis. Compared with vehicle controls, administration of RvE1 resulted in significant improvement of disease activity indices (e.g., body weight, colon length) concomitant with increased ALPI expression in the intestinal epithelium. Moreover, inhibition of ALPI activity resulted in increased severity of colitis in DSS-treated animals and partially abrogated the protective influence of RvE1. Together, these data implicate a previously unappreciated role for ALPI in RvE1-mediated inflammatory resolution.


Asunto(s)
Fosfatasa Alcalina/genética , Ácido Eicosapentaenoico/análogos & derivados , Inflamación/prevención & control , Mucosa Intestinal/enzimología , Lipopolisacáridos/antagonistas & inhibidores , Animales , Colitis/prevención & control , Ácido Eicosapentaenoico/farmacología , Células Epiteliales/química , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Receptores de Superficie Celular/análisis , Activación Transcripcional
3.
J Exp Med ; 198(7): 999-1010, 2003 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-14530374

RESUMEN

Neutrophil migration across mucosal epithelium during inflammatory episodes involves the precise orchestration of a number a cell surface molecules and signaling pathways. After successful migration to the apical epithelial surface, apically localized epithelial proteins may serve to retain PMN at the lumenal surface. At present, identification of apical epithelial ligands and their PMN counter-receptors remain elusive. Therefore, to define the existence of apical epithelial cell surface proteins involved in PMN-epithelial interactions, we screened a panel of antibodies directed against epithelial plasma membranes. This strategy identified one antibody (OE-1) that both localized to the apical cell membrane and significantly inhibited PMN transmigration across epithelial monolayers. Microsequence analysis revealed that OE-1 recognized human decay-accelerating factor (DAF, CD55). DAF is a highly glycosylated, 70-80-kD, glycosyl-phosphatidyinositol-linked protein that functions predominantly as an inhibitor of autologous complement lysis. DAF suppression experiments using antisense oligonucleotides or RNA interference revealed that DAF may function as an antiadhesive molecule promoting the release of PMN from the lumenal surface after transmigration. Similarly, peptides corresponding to the antigen recognition domain of OE-1 resulted in accumulation of PMN on the apical epithelial surface. The elucidation of DAF as an apical epithelial ligand for PMN provides a target for novel anti-inflammatory therapies directed at quelling unwanted inflammatory episodes.


Asunto(s)
Antígenos CD55/fisiología , Neutrófilos/fisiología , Secuencia de Aminoácidos , Sitios de Unión , Antígenos CD55/química , Adhesión Celular , Movimiento Celular , Células Cultivadas , Células Epiteliales/fisiología , Humanos , Datos de Secuencia Molecular , Membrana Mucosa/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA