Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
1.
PLoS One ; 8(3): e59418, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555668

RESUMEN

Cell-in-cell structures refer to a unique phenomenon that one living cell enters into another living cell intactly, occurring between homotypic tumor cells or tumor (or other tissue cells) and immune cells (named as heterotypic cell-in-cell structure). In the present study, through a large scale of survey we observed that heterotypic cell-in-cell structure formation occurred commonly in vitro with host cells derived from different human carcinomas as well as xenotypic mouse tumor cell lines. Most of the lineages of human immune cells, including T, B, NK cells, monocytes as well as in vitro activated LAK cells, were able to invade tumor cell lines. Poorly differentiated stem cells were capable of internalizing immune cells as well. More significantly, heterotypic tumor/immune cell-in-cell structures were observed in a higher frequency in tumor-derived tissues than those in adjacent tissues. In mouse hepatitis models, heterotypic immune cell/hepatocyte cell-in-cell structures were also formed in a higher frequency than in normal controls. After in vitro culture, different forms of internalized immune cells in heterotypic cell-in-cell structures were observed, with one or multiple immune cells inside host cells undergoing resting, degradation or mitosis. More strikingly, some internalized immune cells penetrated directly into the nucleus of target cells. Multinuclear cells with aneuploid nucleus were formed in target tumor cells after internalizing immune cells as well as in situ tumor regions. Therefore, with the prevalence of heterotypic cell-in-cell structures observed, we suggest that shielding of immune cells inside tumor or inflammatory tissue cells implies the formation of aneuploidy with the increased multinucleation as well as fine-tuning of microenvironment under pathological status, which may define distinct mechanisms to influence the etiology and progress of tumors.


Asunto(s)
Aneuploidia , Carcinoma/patología , Comunicación Celular/inmunología , Transformación Celular Neoplásica/patología , Células Gigantes/patología , Neoplasias/patología , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Carcinoma/inmunología , Diferenciación Celular , Transformación Celular Neoplásica/inmunología , Células Gigantes/inmunología , Hepatocitos/inmunología , Hepatocitos/patología , Humanos , Células Asesinas Activadas por Linfocinas/inmunología , Células Asesinas Activadas por Linfocinas/patología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Ratones , Mitosis , Neoplasias/inmunología , Células Madre/inmunología , Células Madre/patología , Linfocitos T/inmunología , Linfocitos T/patología , Células Tumorales Cultivadas
2.
Brain Pathol ; 21(4): 389-404, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21083634

RESUMEN

Transmigration of neutrophil [polymorphonuclear neutrophil (PMN)] across the blood-brain barrier (BBB) is a critical event in the pathogenesis of bacterial meningitis. We have shown that IbeA is able to induce meningitic Escherichia coli invasion of brain microvascular endothelial cells (BMECs), which constitutes the BBB. In this report, we provide evidence that IbeA and its receptor, vimentin, play a key role in E. coli-induced PMN transmigration across BMEC. In vitro and in vivo studies indicated that the ibeA-deletion mutant ZD1 was significantly less active in stimulating PMN transmigration than the parent strain E44. ZD1 was fully complemented by the ibeA gene and its product. E. coli-induced PMN transmigration was markedly inhibited by withaferin A, a dual inhibitor of vimentin and proteasome. These cellular effects were significantly stimulated and blocked by overexpression of vimentin and its head domain deletion mutant in human BMEC, respectively. Our studies further demonstrated that IbeA-induced PMN migration was blocked by bortezomib, a proteasomal inhibitor and correlated with upregulation of endothelial ICAM-1 and CD44 expression through proteasomal regulation of NFκB activity. Taken together, our data suggested that IbeA and vimentin contribute to E. coli K1-stimulated PMN transendothelial migration that is correlated with upregulation of adhesion molecule expression at the BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Movimiento Celular , Células Endoteliales/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de la Membrana/metabolismo , Meningitis por Escherichia coli/metabolismo , Neutrófilos/metabolismo , Migración Transendotelial y Transepitelial/fisiología , Animales , Células Cultivadas , Células Endoteliales/citología , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Meningitis por Escherichia coli/inmunología , Ratones , Neutrófilos/inmunología , Neutrófilos/microbiología , Vimentina/inmunología , Vimentina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA