Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncoimmunology ; 7(2): e1387347, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29308312

RESUMEN

Converting an immunosuppressive melanoma microenvironment into one that favors the induction of antitumor immunity is indispensable for effective cancer immunotherapy. In the current study we demonstrate that oat-derived ß-(1-3)-(1-4)-glucan of 200 kDa molecular size (BG34-200) previously shown to mediate direct interaction with macrophages could alter the immune signature within melanoma microenvironment. Systemic administration of BG34-200 resulted in reversion of tolerant melanoma microenvironment to an immunogenic one that allows M1-type activation of macrophages, the induction of pro-inflammatory cytokines/chemokines including IFN-γ, TNF-α, CXCL9, and CXCL10, and enhanced IRF1 and PD-L1 expression. In turn, BG34-200 induced a superior antitumor response against primary and lung metastatic B16F10 melanoma compared to untreated controls. The enhanced tumor destruction was accompanied with significantly increased tumor infiltration of CD4+ and CD8+ T cells as well as elevated IFN-γ in the tumor sites. Systemic administration of BG34-200 also provoked systemic activation of tumor draining lymph node T cells that recognize antigens naturally expressing in melanoma (gp100/PMEL). Mechanistic studies using CD11b-knockout (KO), CD11 c-DTR transgenic mice and nude mice revealed that macrophages, DCs, T cells and NK cells were all required for the BG34-200-induced therapeutic benefit. Studies using IFN-γ-KO transgenic mice showed that IFN-γ was essential for the BG34-200-elicited antitumor response. Beyond melanoma, the therapeutic efficacy of BG34-200 and its immune stimulating activity were demonstrated in a mouse model of osteosarcoma. Together, BG34-200 is highly effective in modulating antitumor immunity. Our data support the potential therapeutic use of this novel immune modulator in the treatment of metastatic melanoma.

2.
Front Immunol ; 8: 1390, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29109732

RESUMEN

Lymph node (LN) plays a critical role in tumor cell survival outside of the primary tumor sites and dictates overall clinical response in many tumor types (1, 2). Previously, we and others have demonstrated that CCL3 plays an essential role in orchestrating T cell-antigen-presenting cell (APC) encounters in the draining LN following vaccination, and such interactions enhance the magnitude of the memory T cell pool (3-5). In the current study, we investigate the cellular responses in the tumor-draining lymph nodes (TDLNs) of a CCL3-secreting CT26 colon tumor (L3TU) as compared to wild-type tumor (WTTU) during the priming phase of an antitumor response (≤10 days). In comparison to WTTU, inoculation of L3TU resulted in suppressed tumor growth, a phenomenon that is accompanied by altered in vivo inflammatory responses on several fronts. Autologous tumor-derived CCL3 (aCCL3) secretion by L3TU bolstered the recruitment of T- and B-lymphocytes, tissue-migratory CD103+ dendritic cells (DCs), and CD49b+ natural killer (NK) cells, resulting in significant increases in the differentiation and activation of multiple Interferon-gamma (IFNγ)-producing leukocytes in the TDLN. During this early phase of immune priming, NK cells constitute the major producers of IFNγ in the TDLN. CCL3 also enhances CD8+ T cell proliferation and differentiation by augmenting DC capacity to drive T cell activation in the TDLN. Our results revealed that CCL3-dependent IFNγ production and CCL3-induced DC maturation drive the priming of effective antitumor immunity in the TDLN.

3.
Science ; 353(6297): 399-403, 2016 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-27463676

RESUMEN

Cancers often evade immune surveillance by adopting peripheral tissue- tolerance mechanisms, such as the expression of programmed cell death ligand 1 (PD-L1), the inhibition of which results in potent antitumor immunity. Here, we show that cyclin-dependent kinase 5 (Cdk5), a serine-threonine kinase that is highly active in postmitotic neurons and in many cancers, allows medulloblastoma (MB) to evade immune elimination. Interferon-γ (IFN-γ)-induced PD-L1 up-regulation on MB requires Cdk5, and disruption of Cdk5 expression in a mouse model of MB results in potent CD4(+) T cell-mediated tumor rejection. Loss of Cdk5 results in persistent expression of the PD-L1 transcriptional repressors, the interferon regulatory factors IRF2 and IRF2BP2, which likely leads to reduced PD-L1 expression on tumors. Our finding highlights a central role for Cdk5 in immune checkpoint regulation by tumor cells.


Asunto(s)
Antígeno B7-H1/genética , Neoplasias Cerebelosas/inmunología , Quinasa 5 Dependiente de la Ciclina/fisiología , Regulación Neoplásica de la Expresión Génica , Meduloblastoma/inmunología , Neoplasias Experimentales/inmunología , Escape del Tumor/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Neoplasias Cerebelosas/genética , Quinasa 5 Dependiente de la Ciclina/genética , Humanos , Vigilancia Inmunológica , Factor 2 Regulador del Interferón/genética , Factor 2 Regulador del Interferón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias Experimentales/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
J Immunol ; 196(9): 3653-64, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26994221

RESUMEN

In inflamed lymph nodes, Ag-specific CD4(+) and CD8(+) T cells encounter Ag-bearing dendritic cells and, together, this complex enhances the release of CCL3 and CCL4, which facilitate additional interaction with naive CD8(+) T cells. Although blocking CCL3 and CCL4 has no effect on primary CD8(+) T cell responses, it dramatically impairs the development of memory CD8(+) T cells upon Ag rechallenge. Despite the absence of detectable surface CCR5 expression on circulating native CD8(+) T cells, these data imply that naive CD8(+) T cells are capable of expressing surface CCR5 prior to cognate Ag-induced TCR signaling in inflamed lymph nodes; however, the molecular mechanisms have not been characterized to date. In this study, we show that CCR5, the receptor for CCL3 and CCL4, can be transiently upregulated on a subset of naive CD8(+) T cells and that this upregulation is dependent on direct contact with the high endothelial venule in inflamed lymph node. Binding of CD62L and CD11a on T cells to their ligands CD34 and CD54 on the high endothelial venule can be enhanced during inflammation. This enhanced binding and subsequent signaling promote the translocation of CCR5 molecules from intracellular vesicles to the surface of the CD8(+) T cell. The upregulation of CCR5 on the surface of the CD8(+) T cells increases the number of contacts with Ag-bearing dendritic cells, which ultimately results in increased CD8(+) T cell response to Ag rechallenge.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Ganglios Linfáticos/inmunología , Receptores CCR5/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Presentación de Antígeno , Antígenos CD34/inmunología , Antígenos CD34/metabolismo , Antígeno CD11a/inmunología , Antígeno CD11a/metabolismo , Células Dendríticas/inmunología , Inflamación , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Selectina L/inmunología , Selectina L/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/patología , Activación de Linfocitos , Ratones , Receptores CCR5/genética , Linfocitos T Colaboradores-Inductores/metabolismo
5.
Cancer Res ; 76(6): 1641-52, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26801976

RESUMEN

More than half of T-cell acute lymphoblastic leukemia (T-ALL) patients harbor gain-of-function mutations in the intracellular domain of Notch1. Diffuse infiltration of the bone marrow commonly occurs in T-ALL and relapsed B-cell acute lymphoblastic leukemia patients, and is associated with worse prognosis. However, the mechanism of leukemia outgrowth in the marrow and the resulting biologic impact on hematopoiesis are poorly understood. Here, we investigated targetable cellular and molecular abnormalities in leukemia marrow stroma responsible for the suppression of normal hematopoiesis using a T-ALL mouse model and human T-ALL xenografts. We found that actively proliferating leukemia cells inhibited normal hematopoietic stem and progenitor cell (HSPC) proliferation and homing to the perivascular region. In addition, leukemia development was accompanied by the suppression of the endosteum-lining osteoblast population. We further demonstrated that aberrant Notch activation in the stroma plays an important role in negatively regulating the expression of CXLC12 on osteoblasts and their differentiation. Notch blockade reversed attenuated HSPC cycling, leukemia-associated abnormal blood lineage distribution, and thrombocytopenia as well as recovered osteoblast and HSPC abundance and improved the hematopoietic-supportive functions of osteoblasts. Finally, we confirmed that reduced osteoblast frequency and enhanced Notch signaling were also features of the marrow stroma of human ALL tissues. Collectively, our findings suggest that therapeutically targeting the leukemia-infiltrated hematopoietic niche may restore HSPC homeostasis and improve the outcome of ALL patients.


Asunto(s)
Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Osteoblastos/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores Notch/metabolismo , Microambiente Tumoral/fisiología , Adolescente , Adulto , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Médula Ósea/patología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Niño , Preescolar , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/patología , Humanos , Lactante , Ratones , Ratones Endogámicos C57BL , Osteoblastos/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Transducción de Señal/fisiología , Adulto Joven
6.
Acta Trop ; 92(3): 193-203, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15533287

RESUMEN

C57BL/6 mice deficient in one or two copies of the gene for tumor necrosis factor alpha (TNF-alpha) were more susceptible to Trypanosoma congolense infection than their resistant, wild-type counterparts. The number of TNF-alpha genes was correlated with the capacity to control parasitaemia and with survival time. Absence of TNF-alpha resulted in a diminished capacity to form germinal centres in lymph nodes and spleen. Since germinal centres are involved in antibody production and affinity maturation, the susceptibility of the TNF-alpha-deficient mice could have been due to this secondary defect. Despite the lack of the germinal centres, the antibody responses to internal and exposed trypanosome antigens and to non-trypanosome antigens were not significantly different. Also the relative avidities measured in infected sera did not significantly differ between the two mouse strains. These data suggest that the role of TNF-alpha in control of T. congolense was not due to its role in the development of an antibody response.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Trypanosoma congolense/inmunología , Tripanosomiasis Africana/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Afinidad de Anticuerpos/inmunología , Susceptibilidad a Enfermedades , Genotipo , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Parasitemia/inmunología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...