Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Natl Cancer Inst ; 107(1): 363, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25505237

ABSTRACT

BACKGROUND: Regulatory T cells (Treg) and tumor-exosomes are thought to play a role in preventing the rejection of malignant cells in patients bearing nasopharyngeal carcinoma (NPC). METHODS: Treg recruitment by exosomes derived from NPC cell lines (C15/C17-Exo), exosomes isolated from NPC patients' plasma (Patient-Exo), and CCL20 were tested in vitro using Boyden chamber assays and in vivo using a xenograft SCID mouse model (n = 5), both in the presence and absence of anti-CCL20 monoclonal antibodies (mAb). Impact of these NPC exosomes (NPC-Exo) on Treg phenotype and function was determined using adapted assays (FACS, Q-PCR, ELISA, and MLR). Experiments were performed in comparison with exosomes derived from plasma of healthy donors (HD-Exo). The Student's t test was used for group comparisons. All statistical tests were two-sided. RESULTS: CCL20 allowed the intratumoral recruitment of human Treg. NPC-Exo also facilitated Treg recruitment (3.30 ± 0.34 fold increase, P < .001), which was statistically significantly inhibited (P < .001) by an anti-CCL20 blocking mAb. NPC-Exo also recruited conventional CD4(+)CD25(-) T cells and mediated their conversion into inhibitory CD4(+)CD25(high) cells. Moreover, NPC-Exo enhanced (P = .0048) the expansion of human Treg, inducing the generation of Tim3(Low) Treg with increased expression of CD25 and FOXP3. Finally, NPC-Exo induced an overexpression of cell markers associated with Treg phenotype, properties and recruitment capacity. For example, GZMB mean fold change was 21.45 ± 1.75 (P < .001). These results were consistent with a stronger suppression of responder cells' proliferation and the secretion of immunosuppressive cytokines (IL10, TGFB1). CONCLUSION: Interactions between NPC-Exo and Treg represent a newly defined mechanism that may be involved in regulating peripheral tolerance by tumors and in supporting immune evasion in human NPC.


Subject(s)
Carcinoma/metabolism , Exosomes/metabolism , Nasopharyngeal Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Carcinoma/immunology , Cell Line, Tumor , Cell Proliferation , Chemokine CCL20/metabolism , Exosomes/immunology , Flow Cytometry , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Interleukin-10/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, SCID , Nasopharyngeal Neoplasms/immunology , Real-Time Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transforming Growth Factor beta1/immunology
2.
Mediators Inflamm ; 2014: 564296, 2014.
Article in English | MEDLINE | ID: mdl-25525301

ABSTRACT

Virus infections are involved in chronic inflammation and, in some cases, cancer development. Although a viral infection activates the immune system's response that eradicates the pathogen mainly through inflammatory mechanisms, it is now recognized that this inflammatory condition is also favorable to the development of tumors. Indeed, it is well described that viruses, such as hepatitis C virus (HCV), Epstein Barr virus (EBV), human papillomavirus (HPV) or human T-cell lymphotropic virus type-1 (HTLV-1), are important risk factors for tumor malignancies. The inflammatory response is a fundamental immune mechanism which involves several molecular and cellular components consisting of cytokines and chemokines that are released by various proinflammatory cells. In parallel to this process, some endogenous recruited components release anti-inflammatory mediators to restore homeostasis. The development of tools and strategies using viruses to hijack the immune response is mostly linked to the presence of regulatory T-cells (Treg) that can inhibit inflammation and antiviral responses of other effector cells. In this review, we will focus on current understanding of the role of natural and induced Treg in the control and the resolution of inflammatory response in HCV-, HTLV-1-, and EBV-associated cancers.


Subject(s)
Hepacivirus/metabolism , Herpesvirus 4, Human/metabolism , Human T-lymphotropic virus 1/metabolism , Inflammation/metabolism , Neoplasms/virology , T-Lymphocytes, Regulatory/cytology , Animals , Carcinoma/virology , Carcinoma, Hepatocellular/virology , Hodgkin Disease/virology , Homeostasis , Humans , Immunity, Innate , Leukemia, T-Cell/virology , Liver Neoplasms/virology , Lymphoma, T-Cell/virology , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/virology , Risk Factors
3.
Br J Haematol ; 166(6): 875-90, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25041527

ABSTRACT

Epstein-Barr Virus (EBV) is present in the neoplastic cells of around 20-30% of patients with Hodgkin Lymphoma (HL). Although, an immunosuppressive environment is currently described in HL patients, little is known concerning the regulatory mechanism induced by EBV proteins expression in tumour cells. This study aimed to investigate an association between regulatory Type 1 cells (Tr1) and EBV tissue positivity in HL patients. Transcriptomic analysis of both EBV-positive and EBV-negative tumours showed that EBV infection increased gene expression of Tr1-related markers (ITGA2, ITGB2, LAG3) and associated-immunosuppressive cytokines (IL10). This up-regulation was associated with an over-expression of several chemokine markers known to attract T-helper type 2 (Th2) and regulatory T cells thus contributing to immune suppression. This Tr1 cells recruitment in EBV-positive HL was confirmed by immunohistochemical analysis of frozen nodes biopsies and by flow cytometric analysis of peripheral blood mononuclear cells of EBV-positive patients. Additionally, we showed that IL10 production was significantly enhanced in tumours and blood of EBV-positive HL patients. Our results propose a new model in which EBV can recruit Tr1 cells to the nodes' microenvironment, suggesting that the expression of EBV proteins in tumour cells could enable the escape of EBV-infected tumour cells from the virus-specific CTL response.


Subject(s)
Epstein-Barr Virus Infections/immunology , Hodgkin Disease/immunology , T-Lymphocytes, Regulatory/immunology , Adolescent , Adult , Aged , Biomarkers/metabolism , Chemokines/metabolism , Child , Cytokines/metabolism , Female , Gene Expression Regulation, Neoplastic , Hodgkin Disease/virology , Humans , Male , Middle Aged , Phenotype , T-Lymphocytes, Regulatory/virology , Th2 Cells/immunology , Th2 Cells/virology , Up-Regulation , Young Adult
4.
ISRN Hepatol ; 2013: 928485, 2013.
Article in English | MEDLINE | ID: mdl-27335834

ABSTRACT

Hepatitis C virus (HCV) is an important causative agent of liver disease, but factors that determine the resolution or progression of infection are poorly understood. In this study, we suggested that existence of immunosuppressive mechanisms, supported by regulatory T cells and especially the regulatory T cell 1 subset (Tr1), may explain the impaired immune response during infection and thus the fibrosis aggravation to hepatocellular carcinoma (HCC). Using quantitative real-time PCR, we investigated the intra-hepatic presence of Tr1 cells in biopsies from a genotype 1b infected patient followed for an 18-year period from cirrhosis to HCC. We described a significant increase of gene expression in particular for the cytokines IL-10, TGF-ß, and their receptors that were perfectly correlated with an increased expression of the Tr1 specific markers (combined expression of CD4, CD18, and CD49b). This was strongly marked since the patient evolved in the pathology and could explain the failure of the treatment. In conclusion, evidence of regulatory T cell installation in the liver of chronically infected patient with cirrhosis and HCC suggests for the first time a key role for these cells in the course of HCV infection.

5.
J Immunother ; 35(3): 254-66, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22421943

ABSTRACT

Epstein-Barr virus (EBV) is associated with several malignant diseases that can be distinguished by their patterns of viral latent gene expression. We developed here an original peptidic approach to favor the induction of a specific CD4+ T-cell response against EBV latency II malignancies (Hodgkin's lymphoma, nasopharyngeal carcinoma, T/NK lymphoma). Previously, we selected 6 peptides derived from EBV nuclear antigen-1, latency membrane proteins (LMP)-1, and LMP-2 highly promiscuous for major histocompatibility complex class II molecules and showed their ability to induce interferon-γ-secreting CD4+ T cells. We confirmed here that all peptides used in cocktail are recognized by human CD4+ memory T cells from healthy donors, inducing a broad T-helper (Th)1 cytokine secretion interferon-γ, interleukin-2. Furthermore, we have generated EBV-specific CD4+ T-cell lines and proved their cytotoxic potential, not only on original models expressing latency II antigens (EBV-transformed T cell or monocyte), but also on lymphoblastoid cell lines expressing latency III antigens (lymphoblastoid cell lines). In addition, granzyme B enzyme-linked immunospot assays suggested that a part of this specific cytotoxic activity could be linked to the granule lytic pathway. Very importantly, we have showed that neither phenotypical changes nor functional activities of CD4+CD25+CD127(low)-regulatory T cells were observed in response to EBV+ peptides, avoiding any risk of aggravation of the preexisting immunosuppressive environment reported in EBV-associated malignancies. In conclusion, our promiscuous peptide cocktail could be used safely in immunotherapeutic approaches against EBV latency II malignancies, mainly to prevent relapse in high-risk patients further to classic treatments.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Granzymes/metabolism , Humans , Immunologic Memory , Peptides/chemistry , Th1 Cells/immunology , Th1 Cells/metabolism
6.
PLoS One ; 7(2): e32197, 2012.
Article in English | MEDLINE | ID: mdl-22359669

ABSTRACT

BACKGROUND: H-1 parvovirus (H-1 PV), a rodent autonomous oncolytic parvovirus, has emerged as a novel class of promising anticancer agents, because of its ability to selectively find and destroy malignant cells. However, to probe H-1 PV multimodal antitumor potential one of the major prerequisites is to decipher H-1 PV direct interplay with human immune system, and so prevent any risk of impairment. METHODOLOGY/PRINCIPAL FINDINGS: Non activated peripheral blood mononuclear cells (PBMCs) are not sensitive to H-1 PV cytotoxic effect. However, the virus impairs both activated PBMC proliferation ability and viability. This effect is related to H-1 PV infection as evidenced by Western blotting detection of H-1 PV main protein NS1. However, TCID50 experiments did not allow newly generated virions to be detected. Moreover, flow cytometry has shown that H-1 PV preferentially targets B lymphocytes. Despite seeming harmful at first sight, H-1 PV seems to affect very few NK cells and CD8+ T lymphocytes and, above all, clearly does not affect human neutrophils and one of the major CD4+ T lymphocyte subpopulation. Very interestingly, flow cytometry analysis and ELISA assays proved that it even activates human CD4+ T cells by increasing activation marker expression (CD69 and CD30) and both effective Th1 and Th2 cytokine secretion (IL-2, IFN-γ and IL-4). In addition, H-1 PV action does not come with any sign of immunosuppressive side effect. Finally, we have shown the efficiency of H-1 PV on xenotransplanted human nasopharyngeal carcinoma, in a SCID mouse model reconstituted with human PBMC. CONCLUSIONS/SIGNIFICANCE: Our results show for the first time that a wild-type oncolytic virus impairs some immune cell subpopulations while directly activating a Helper CD4+ T cell response. Thus, our data open numerous gripping perspectives of investigation and strongly argue for the use of H-1 PV as an anticancer treatment.


Subject(s)
Biological Therapy/methods , CD4-Positive T-Lymphocytes/virology , H-1 parvovirus/immunology , Oncolytic Viruses/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Carcinoma , Humans , Mice , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/therapy , T-Lymphocytes, Regulatory , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...