Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(3): 997-1006, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30602457

RESUMEN

Glioblastoma multiforme (GBM) is a highly aggressive malignant brain tumor with fatal outcome. Tumor-associated macrophages and microglia (TAMs) have been found to be major tumor-promoting immune cells in the tumor microenvironment. Hence, modulation and reeducation of tumor-associated macrophages and microglia in GBM is considered a promising antitumor strategy. Resident microglia and invading macrophages have been shown to have distinct origin and function. Whereas yolk sac-derived microglia reside in the brain, blood-derived monocytes invade the central nervous system only under pathological conditions like tumor formation. We recently showed that disruption of the SIRPα-CD47 signaling axis is efficacious against various brain tumors including GBM primarily by inducing tumor phagocytosis. However, most effects are attributed to macrophages recruited from the periphery but the role of the brain resident microglia is unknown. Here, we sought to utilize a model to distinguish resident microglia and peripheral macrophages within the GBM-TAM pool, using orthotopically xenografted, immunodeficient, and syngeneic mouse models with genetically color-coded macrophages (Ccr2RFP) and microglia (Cx3cr1GFP). We show that even in the absence of phagocytizing macrophages (Ccr2RFP/RFP), microglia are effector cells of tumor cell phagocytosis in response to anti-CD47 blockade. Additionally, macrophages and microglia show distinct morphological and transcriptional changes. Importantly, the transcriptional profile of microglia shows less of an inflammatory response which makes them a promising target for clinical applications.


Asunto(s)
Neoplasias Encefálicas/inmunología , Antígeno CD47/inmunología , Glioblastoma/inmunología , Microglía/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/inmunología , Fagocitosis , Receptores Inmunológicos/inmunología , Transducción de Señal/inmunología , Animales , Neoplasias Encefálicas/patología , Antígeno CD47/genética , Glioblastoma/genética , Glioblastoma/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Microglía/patología , Monocitos/inmunología , Monocitos/patología , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Receptores Inmunológicos/genética , Transducción de Señal/genética
2.
PLoS One ; 11(4): e0153550, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27092773

RESUMEN

Tumor-associated macrophages (TAMs) represent an important cellular subset within the glioblastoma (WHO grade IV) microenvironment and are a potential therapeutic target. TAMs display a continuum of different polarization states between antitumorigenic M1 and protumorigenic M2 phenotypes, with a lower M1/M2 ratio correlating with worse prognosis. Here, we investigated the effect of macrophage polarization on anti-CD47 antibody-mediated phagocytosis of human glioblastoma cells in vitro, as well as the effect of anti-CD47 on the distribution of M1 versus M2 macrophages within human glioblastoma cells grown in mouse xenografts. Bone marrow-derived mouse macrophages and peripheral blood-derived human macrophages were polarized in vitro toward M1 or M2 phenotypes and verified by flow cytometry. Primary human glioblastoma cell lines were offered as targets to mouse and human M1 or M2 polarized macrophages in vitro. The addition of an anti-CD47 monoclonal antibody led to enhanced tumor-cell phagocytosis by mouse and human M1 and M2 macrophages. In both cases, the anti-CD47-induced phagocytosis by M1 was more prominent than that for M2. Dissected tumors from human glioblastoma xenografted within NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice and treated with anti-CD47 showed a significant increase of M1 macrophages within the tumor. These data show that anti-CD47 treatment leads to enhanced tumor cell phagocytosis by both M1 and M2 macrophage subtypes with a higher phagocytosis rate by M1 macrophages. Furthermore, these data demonstrate that anti-CD47 treatment alone can shift the phenotype of macrophages toward the M1 subtype in vivo.


Asunto(s)
Anticuerpos/farmacología , Antígeno CD47/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Fagocitosis/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Glioblastoma/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos NOD , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA