Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Cell Rep ; 34(13): 108917, 2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33789113

RESUMEN

Tumor-associated macrophages (TAMs) play an important role in tumor immunity and comprise of subsets that have distinct phenotype, function, and ontology. Transcriptomic analyses of human medulloblastoma, the most common malignant pediatric brain cancer, showed that medulloblastomas (MBs) with activated sonic hedgehog signaling (SHH-MB) have significantly more TAMs than other MB subtypes. Therefore, we examined MB-associated TAMs by single-cell RNA sequencing of autochthonous murine SHH-MB at steady state and under two distinct treatment modalities: molecular-targeted inhibitor and radiation. Our analyses reveal significant TAM heterogeneity, identify markers of ontologically distinct TAM subsets, and show the impact of brain microenvironment on the differentiation of tumor-infiltrating monocytes. TAM composition undergoes dramatic changes with treatment and differs significantly between molecular-targeted and radiation therapy. We identify an immunosuppressive monocyte-derived TAM subset that emerges with radiation therapy and demonstrate its role in regulating T cell and neutrophil infiltration in MB.


Asunto(s)
Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/terapia , Proteínas Hedgehog/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Meduloblastoma/patología , Meduloblastoma/terapia , Animales , Linfocitos T CD8-positivos/inmunología , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/inmunología , Marcadores Genéticos , Humanos , Meduloblastoma/genética , Meduloblastoma/inmunología , Ratones , Microglía/patología , Monocitos/patología , Análisis de la Célula Individual , Transcripción Genética , Microambiente Tumoral
3.
Cell Rep ; 31(13): 107825, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32610126

RESUMEN

Activated macrophages must carefully calibrate their inflammatory responses to balance efficient pathogen control with inflammation-mediated tissue damage, but the molecular underpinnings of this "balancing act" remain unclear. Using genetically engineered mouse models and primary macrophage cultures, we show that Toll-like receptor (TLR) signaling induces the expression of the transcription factor Spic selectively in patrolling monocytes and tissue macrophages by a nuclear factor κB (NF-κB)-dependent mechanism. Functionally, Spic downregulates pro-inflammatory cytokines and promotes iron efflux by regulating ferroportin expression in activated macrophages. Notably, interferon-gamma blocks Spic expression in a STAT1-dependent manner. High levels of interferon-gamma are indicative of ongoing infection, and in its absence, activated macrophages appear to engage a "default" Spic-dependent anti-inflammatory pathway. We also provide evidence for the engagement of this pathway in sterile inflammation. Taken together, our findings uncover a pathway wherein counter-regulation of Spic by NF-κB and STATs attune inflammatory responses and iron metabolism in macrophages.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Inflamación/metabolismo , Inflamación/patología , Hierro/metabolismo , Macrófagos/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Animales , Transporte Biológico , Regulación hacia Abajo/genética , Femenino , Hemo/metabolismo , Interferón gamma/metabolismo , Ligandos , Activación de Macrófagos , Masculino , Ratones Endogámicos C57BL , Monocitos/metabolismo , Receptores Toll-Like/metabolismo
4.
Cell ; 180(6): 1098-1114.e16, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32169218

RESUMEN

The immunosuppressive tumor microenvironment (TME) is a major barrier to immunotherapy. Within solid tumors, why monocytes preferentially differentiate into immunosuppressive tumor-associated macrophages (TAMs) rather than immunostimulatory dendritic cells (DCs) remains unclear. Using multiple murine sarcoma models, we find that the TME induces tumor cells to produce retinoic acid (RA), which polarizes intratumoral monocyte differentiation toward TAMs and away from DCs via suppression of DC-promoting transcription factor Irf4. Genetic inhibition of RA production in tumor cells or pharmacologic inhibition of RA signaling within TME increases stimulatory monocyte-derived cells, enhances T cell-dependent anti-tumor immunity, and synergizes with immune checkpoint blockade. Furthermore, an RA-responsive gene signature in human monocytes correlates with an immunosuppressive TME in multiple human tumors. RA has been considered as an anti-cancer agent, whereas our work demonstrates its tumorigenic capability via myeloid-mediated immune suppression and provides proof of concept for targeting this pathway for tumor immunotherapy.


Asunto(s)
Monocitos/inmunología , Tretinoina/metabolismo , Microambiente Tumoral/inmunología , Animales , Carcinogénesis/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Humanos , Terapia de Inmunosupresión/métodos , Inmunoterapia/métodos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo
5.
Nature ; 569(7754): 73-78, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30996346

RESUMEN

Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) are pathologically activated neutrophils that are crucial for the regulation of immune responses in cancer. These cells contribute to the failure of cancer therapies and are associated with poor clinical outcomes. Despite recent advances in the understanding of PMN-MDSC biology, the mechanisms responsible for the pathological activation of neutrophils are not well defined, and this limits the selective targeting of these cells. Here we report that mouse and human PMN-MDSCs exclusively upregulate fatty acid transport protein 2 (FATP2). Overexpression of FATP2 in PMN-MDSCs was controlled by granulocyte-macrophage colony-stimulating factor, through the activation of the STAT5 transcription factor. Deletion of FATP2 abrogated the suppressive activity of PMN-MDSCs. The main mechanism of FATP2-mediated suppressive activity involved the uptake of arachidonic acid and the synthesis of prostaglandin E2. The selective pharmacological inhibition of FATP2 abrogated the activity of PMN-MDSCs and substantially delayed tumour progression. In combination with checkpoint inhibitors, FATP2 inhibition blocked tumour progression in mice. Thus, FATP2 mediates the acquisition of immunosuppressive activity by PMN-MDSCs and represents a target to inhibit the functions of PMN-MDSCs selectively and to improve the efficiency of cancer therapy.


Asunto(s)
Proteínas de Transporte de Ácidos Grasos/metabolismo , Ácidos Grasos/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Neutrófilos/metabolismo , Anciano , Animales , Ácido Araquidónico/metabolismo , Dinoprostona/metabolismo , Proteínas de Transporte de Ácidos Grasos/antagonistas & inhibidores , Femenino , Humanos , Metabolismo de los Lípidos , Lípidos , Masculino , Ratones , Persona de Mediana Edad , Neutrófilos/patología , Factor de Transcripción STAT5/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...