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.
Adv Exp Med Biol ; 1231: 79-96, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32060848

RESUMEN

T cell-mediated elimination of malignant cells is one cornerstone of endogenous and therapeutically induced antitumor immunity. Tumors exploit numerous regulatory mechanisms to suppress T cell immunity. Regulatory T cells (T regs) play a crucial role in this process due to their ability to inhibit antitumoral immune responses and they are known to accumulate in various cancer entities. The chemokine CCL22, predominately produced by dendritic cells (DCs), regulates T reg migration via binding to its receptor CCR4. CCL22 controls T cell immunity, both by recruiting T regs to the tumor tissue and by promoting the formation of DC-T reg contacts in the lymph node. Here, we review the current knowledge on the role of CCL22 in cancer immunity. After revising the principal mechanisms of CCL22-induced immune suppression, we address the factors leading to CCL22 expression and ways of targeting this chemokine therapeutically. Therapeutic interventions to the CCL22-CCR4 axis may represent a promising strategy in cancer immunotherapy.


Asunto(s)
Quimiocina CCL22/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Transducción de Señal , Microambiente Tumoral , Animales , Humanos , Neoplasias/terapia , Receptores CCR4/metabolismo , Linfocitos T Reguladores/inmunología
2.
J Exp Med ; 216(5): 1170-1181, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-30910796

RESUMEN

Chemokines have crucial roles in organ development and orchestration of leukocyte migration. The chemokine CCL22 is expressed constitutively at high levels in the lymph node, but the functional significance of this expression is so far unknown. Studying a newly established CCL22-deficient mouse, we demonstrate that CCL22 expression by dendritic cells (DCs) promotes the formation of cell-cell contacts and interaction with regulatory T cells (T reg) through their CCR4 receptor. Vaccination of CCL22-deficient mice led to excessive T cell responses that were also observed when wild-type mice were vaccinated using CCL22-deficient DCs. Tumor-bearing mice with CCL22 deficiency showed prolonged survival upon vaccination, and further, CCL22-deficient mice had increased susceptibility to inflammatory disease. In conclusion, we identify the CCL22-CCR4 axis as an immune checkpoint that is crucial for the control of T cell immunity.


Asunto(s)
Células de la Médula Ósea/inmunología , Comunicación Celular/inmunología , Quimiocina CCL22/inmunología , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Línea Celular Tumoral , Movimiento Celular , Quimiocina CCL22/genética , Células HEK293 , Humanos , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR4/metabolismo , Trasplante Homólogo
3.
Cell Death Dis ; 10(2): 116, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30741933

RESUMEN

Ewing sarcoma (EwS) is an aggressive cancer characterized by chromosomal translocations generating fusions of the EWSR1 gene with ETS transcription factors (in 85% FLI1). EWSR1-FLI1 induces gene expression via binding to enhancer-like GGAA-microsatellites, whose activity correlates with the number of consecutive GGAA-repeats. Herein we investigate the role of the secretory neuropeptide CALCB (calcitonin-related polypeptide ß) in EwS, which signals via the CGRP (calcitonin gene-related peptide) receptor complex, containing RAMP1 (receptor activity modifying protein 1) as crucial part for receptor specificity. Analysis of 2678 gene expression microarrays comprising 50 tumor entities and 71 normal tissue types revealed that CALCB is specifically and highly overexpressed in EwS. Time-course knockdown experiments showed that CALCB expression is tightly linked to that of EWSR1-FLI1. Consistently, gene set enrichment analyses of genes whose expression in primary EwS is correlated to that of CALCB indicated that it is co-expressed with other EWSR1-FLI1 target genes and associated with signatures involved in stemness and proliferation. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) data for FLI1 and histone marks from EwS cell lines demonstrated that EWSR1-FLI1 binds to a GGAA-microsatellite close to CALCB, which exhibits characteristics of an active enhancer. Reporter assays confirmed the strong EWSR1-FLI1- and length-dependent enhancer activity of this GGAA-microsatellite. Mass spectrometric analyses of EwS cell culture supernatants demonstrated that CALCB is secreted by EwS cells. While short-term RNA interference-mediated CALCB knockdown had no effect on proliferation and clonogenic growth of EwS cells in vitro, its long-term knockdown decreased EwS growth in vitro and in vivo. Similarly, knockdown of RAMP1 reduced clonogenic/spheroidal growth and tumorigenicity, and small-molecule inhibitors directed against the RAMP1-comprising CGRP receptor reduced growth of EwS. Collectively, our findings suggest that CALCB is a direct EWSR1-FLI1 target and that targeting the CALCB/RAMP1 axis may offer a new therapeutic strategy for inhibition of EwS growth.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/metabolismo , Proteína 1 Modificadora de la Actividad de Receptores/metabolismo , Sarcoma de Ewing/patología , Animales , Péptido Relacionado con Gen de Calcitonina/antagonistas & inhibidores , Péptido Relacionado con Gen de Calcitonina/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos NOD , Repeticiones de Microsatélite/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/uso terapéutico , Proteína 1 Modificadora de la Actividad de Receptores/antagonistas & inhibidores , Proteína 1 Modificadora de la Actividad de Receptores/genética , Sarcoma de Ewing/tratamiento farmacológico , Sarcoma de Ewing/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Trasplante Heterólogo
4.
Oncoimmunology ; 5(9): e1175794, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27757295

RESUMEN

In cancer patients, immunosuppression through regulatory T cells (Treg) is a crucial component of tumor immune evasion and contributes to disease progression. Tumor-infiltrating Treg in particular suppress local effector T cell responses and are associated with poor prognosis in tumors such as human pancreatic cancer or hepatocellular carcinoma (HCC). The chemokine CCL22 is known to recruit Treg into the tumor tissue and many types of human tumors are known to express high levels of CCL22. The mechanisms leading to intratumoral secretion of CCL22 are so far unknown. We demonstrate here that intratumoral CCL22 is induced in tumor-infiltrating immune cells through cancer cell-derived interleukin-1 (IL-1α). In pancreatic cancer and HCC, CCL22 is produced by intratumoral dendritic cells, while the cancer cells themselves do not secrete CCL22 in vitro and in vivo. Incubation of human peripheral blood mononuclear cells (PBMC) or murine splenocytes with tumor cells or tumor cell supernatants strongly induced CCL22 secretion in vitro. Tumor cell supernatants contained IL-1 and CCL22 induction in PBMC could be specifically prevented by the IL-1 receptor antagonist anakinra or by transfection of tumor cell lines with IL-1 siRNA, leading to a suppression of Treg migration. In conclusion, we identify here tumor cell-derived IL-1α as a major inducer of the Treg attracting chemokine CCL22 in human cancer cells. Therapeutic blockade of the IL-1 pathway could represent a promising strategy to inhibit tumor-induced immunosuppression.

6.
Cancer Res ; 75(21): 4483-93, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26432403

RESUMEN

The chemokine CCL22 is abundantly expressed in many types of cancer and is instrumental for intratumoral recruitment of regulatory T cells (Treg), an important subset of immunosuppressive and tumor-promoting lymphocytes. In this study, we offer evidence for a generalized strategy to blunt Treg activity that can limit immune escape and promote tumor rejection. Activation of innate immunity with Toll-like receptor (TLR) or RIG-I-like receptor (RLR) ligands prevented accumulation of Treg in tumors by blocking their immigration. Mechanistic investigations indicated that Treg blockade was a consequence of reduced intratumoral CCL22 levels caused by type I IFN. Notably, stable expression of CCL22 abrogated the antitumor effects of treatment with RLR or TLR ligands. Taken together, our findings argue that type I IFN blocks the Treg-attracting chemokine CCL22 and thus helps limit the recruitment of Treg to tumors, a finding with implications for cancer immunotherapy.


Asunto(s)
Movimiento Celular/inmunología , Quimiocina CCL22/metabolismo , Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Línea Celular Tumoral , Células Dendríticas/inmunología , Progresión de la Enfermedad , Femenino , Humanos , Células Jurkat , Activación de Linfocitos/inmunología , Células MCF-7 , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Reguladores/trasplante , Escape del Tumor/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...