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











Intervalo de año de publicación
1.
J Immunol ; 203(6): 1665-1674, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31434708

RESUMEN

EBV infects and immortalizes B cells in vitro and in vivo. It is the causative agent of most immune deficiency-related lymphoproliferative disorders and is associated with various lymphomas. EBV latency III-transformed B cells are known to express two immunosuppressive molecules, IL-10 and PD-L1, two characteristics of regulatory B cells (Bregs). In this study, we show that, in addition to secretion of the Breg immunosuppressive cytokines IL-10, IL-35, and TGF-ß1, EBV latency III-transformed B cells were able to repress proliferation of their autologous T cells preactivated by CD2, CD3, and CD28. This inhibitory effect was likely caused by CD4+ T cells because EBV latency III-transformed B cells induced a strong proliferation of isolated autologous CD8 T cells. Indeed, EBV was able to promote expansion of autologous FOXP3+ CD39high CTLA4+, Helios+, GITR+, LAG3+ CD4 T cells (i.e., regulatory T cells [Tregs]). Two types of Tregs were induced: unconventional CD25neg and conventional CD25pos Tregs. These Tregs expressed both the latency-associated peptide (LAP) and the PD-1 receptor, two markers of functional Tregs. Expansion of both Treg subtypes depended on PD-L1, whose expression was under the control of LMP1, the main EBV oncogene. These results demonstrate that, like Bregs, EBV latency III-transformed B cells exhibit strong immunoregulatory properties. These data provide clues to the understanding of how after EBV primo-infection, EBV-proliferating B cells can survive in an aggressive immunological environment and later emerge to give rise to EBV-associated B cell lymphomas such as in elderly patients.


Asunto(s)
Linfocitos B/inmunología , Antígeno B7-H1/inmunología , Herpesvirus Humano 4/inmunología , Linfocitos T Reguladores/inmunología , Latencia del Virus/inmunología , Antígenos CD/inmunología , Apirasa/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/inmunología , Línea Celular , Factores de Transcripción Forkhead/inmunología , Proteína Relacionada con TNFR Inducida por Glucocorticoide/inmunología , Humanos , Subunidad alfa del Receptor de Interleucina-2/inmunología
2.
Haematologica ; 102(5): 883-894, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28232371

RESUMEN

While c-Myc dysregulation is constantly associated with highly proliferating B-cell tumors, nuclear factor (NF)-κB addiction is found in indolent lymphomas as well as diffuse large B-cell lymphomas, either with an activated B-cell like phenotype or associated with the Epstein-Barr virus. We raised the question of the effect of c-Myc in B cells with NF-κB activated by three different inducers: Epstein-Barr virus-latency III program, TLR9 and CD40. Induction of c-Myc overexpression increased proliferation of Epstein-Barr virus-latency III immortalized B cells, an effect that was dependent on NF-κB. Results from transcriptomic signatures and functional studies showed that c-Myc overexpression increased Epstein-Barr virus-latency III-driven proliferation depending on NF-κB. In vitro, induction of c-Myc increased proliferation of B cells with TLR9-dependant activation of MyD88, with decreased apoptosis. In the transgenic λc-Myc mouse model with c-Myc overexpression in B cells, in vivo activation of MyD88 by TLR9 induced splenomegaly related to an increased synthesis phase (S-phase) entry of B cells. Transgenic mice with both continuous CD40 signaling in B cells and the λc-Myc transgene developed very aggressive lymphomas with characteristics of activated diffuse large B-cell lymphomas. The main characteristic gene expression profile signatures of these tumors were those of proliferation and energetic metabolism. These results suggest that c-Myc is an NF-κB co-transforming event in aggressive lymphomas with an activated phenotype, activated B-cell like diffuse large B-cell lymphomas. This would explain why NF-κB is associated with both indolent and aggressive lymphomas, and opens new perspectives on the possibility of combinatory therapies targeting both the c-Myc proliferating program and NF-κB activation pathways in diffuse large B-cell lymphomas.


Asunto(s)
Linfocitos B/metabolismo , Transformación Celular Viral , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Apoptosis/genética , Linfocitos B/virología , Antígenos CD40/genética , Antígenos CD40/metabolismo , Línea Celular Transformada , Proliferación Celular/genética , Perfilación de la Expresión Génica/métodos , Herpesvirus Humano 4/fisiología , Humanos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/metabolismo , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal/genética
3.
J Immunol ; 189(1): 181-90, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22661084

RESUMEN

EBV-immortalized B cells induce a complex immune response such that the virus persists as a clinically silent infection for the lifetime of the infected host. B7-H1, also called PD-L1, is a cosignaling molecule of the B7 family that can inhibit activated T cell effectors by interaction with its receptor PD-1. In this work, we have studied the dependence of B7-H1 on NF-κB and c-Myc, the two main transcription factors in EBV latency III proliferating B cells, on various lymphoblastoid and Burkitt lymphoma cell lines, some of them being inducible or not for the EBV latency III program and/or for c-Myc. We found that B7-H1 repressed killing of EBV-immortalized B cells by their autologous T and NK cells. At the mRNA level, NF-κB was a weak inducer whereas c-Myc was a strong repressor of B7-H1 expression, an effect mediated by STAT1 inhibition. At the protein level, B7-H1 molecules were stored in both degradative and unconventional secretory lysosomes. Surface membrane B7-H1 molecules were constitutively internalized and proteolyzed in lysosomes. The EBV latency III program increased the amounts of B7-H1-containing secretory lysosomes and their export to the surface membrane. By repressing actin polymerization, c-Myc blocked secretory lysosome migration and B7-H1 surface membrane export. In addition to B7-H1, various immunoregulatory molecules participating in the immunological synapse are stored in secretory lysosomes. By playing on actin polymerization, c-Myc could thus globally regulate the immunogenicity of transformed B cells, acting on export of secretory lysosomes to plasma membrane.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Antígeno B7-H1/fisiología , Herpesvirus Humano 4/inmunología , Células Asesinas Naturales/inmunología , Lisosomas/metabolismo , Proteínas Proto-Oncogénicas c-myc/fisiología , ARN Viral/fisiología , Subgrupos de Linfocitos T/inmunología , Latencia del Virus/inmunología , Subgrupos de Linfocitos B/patología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/biosíntesis , Transporte Biológico Activo/inmunología , Muerte Celular/inmunología , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Supervivencia Celular/inmunología , Regulación hacia Abajo/inmunología , Humanos , Lisosomas/inmunología , ARN Mensajero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA